CN112574208B - Substituted fused tricyclic derivatives, compositions and uses thereof - Google Patents

Substituted fused tricyclic derivatives, compositions and uses thereof Download PDF

Info

Publication number
CN112574208B
CN112574208B CN202011009604.4A CN202011009604A CN112574208B CN 112574208 B CN112574208 B CN 112574208B CN 202011009604 A CN202011009604 A CN 202011009604A CN 112574208 B CN112574208 B CN 112574208B
Authority
CN
China
Prior art keywords
alkyl
exon
mutation
compound
egfr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202011009604.4A
Other languages
Chinese (zh)
Other versions
CN112574208A (en
Inventor
王义汉
邢青峰
艾义新
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Priority to CN202210996465.1A priority Critical patent/CN115181104A/en
Publication of CN112574208A publication Critical patent/CN112574208A/en
Application granted granted Critical
Publication of CN112574208B publication Critical patent/CN112574208B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The invention provides a substituted fused tricyclic derivative, a composition containing the compound and application of the compound, wherein the substituted fused tricyclic derivative is a compound shown in a formula (I) or a tautomer, a stereoisomer, a prodrug, a crystal form, a pharmaceutically acceptable salt, a hydrate or a solvate thereof. The compounds and compositions of the invention are useful for the treatment and/or prevention of mutant EGFR kinase-mediated tumors, as well as wild-type and/or mutant HER2 kinase-mediated tumors.

Description

Substituted fused tricyclic derivatives, compositions and uses thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to substituted fused tricyclic derivatives with an inhibitory effect on mutant Epidermal Growth Factor Receptor (EGFR) and wild and/or mutant HER2, pharmaceutical compositions containing the same, and preparation methods and applications of the same.
Background
EGFR is a receptor tyrosine kinase that exerts its physiological function in normal tissues by binding to epidermal growth factor (hereinafter also referred to as EGF) as a ligand, and contributes to growth and apoptosis inhibition of epithelial tissues. Furthermore, somatic mutations in the EGFR gene are known to be oncogenes: for example, EGFR lacking 746 th to 750 th amino acids in the exon 19 region (hereinafter also referred to as "exon 19 deletion mutation") and EGFR lacking the leucine th to arginine in the exon 21 region (hereinafter also referred to as "L858R mutation") continuously induce EGF-independent kinase activity and cause growth and survival of cancer cells. For example, these mutations are observed in about 30% -50% of non-small cell lung cancers in east asia, and also in about 10% of non-small cell lung cancers in europe and the united states, and thus are considered to be one of the causes of cancer.
Therefore, research and development of EGFR inhibitors as antitumor agents have been actively conducted and applied to the treatment of EGFR mutation-positive lung cancer. For example, gefitinib, erlotinib and afatinib have high antitumor effects on exon 19 deletion mutants and L858R mutant EGFR-positive lung cancer, but their administration at therapeutic doses causes side effects such as digestive tract diseases and skin diseases, which are generally considered to be attributable to wild-type EGFR.
Recent studies have found that EGFR of some cancers has a novel mutation in which one or more amino acids are inserted into the exon 20 region (hereinafter also referred to as "exon 20 insertion mutation"), and these cancers have low sensitivity to previously known EGFR inhibitors.
On the other hand, a number of rare EGFR mutations, such as point mutations or deletion mutations of exon 18 and point mutations of exon 21, have been reported. For example, a new EGFR point mutation lung cancer has been found in which the glycine encoded by codon 719 in exon 18 has been substituted with an arbitrary amino acid (hereinafter referred to as the G719X mutation) and the leucine encoded by codon 861 in exon 21 has been substituted with glutamine (hereinafter referred to as the L861Q mutation).
HER2 (also known as ErbB2) is a receptor tyrosine kinase belonging to the ErbB2 family.
HER2 is considered to be a protooncogene, and gene amplification, mutation, overexpression, and the like of HER2 have been reported in various cancers. It has been reported that these cancer cells, which are accompanied by the genetic abnormality and overexpression of HER2, have enhanced survival and proliferation signals of cancer cells due to signal activation of HER2 and downstream pathways.
Therefore, it is presumed that an inhibitor capable of controlling the kinase activity of HER2 exerts an antitumor effect by inhibiting HER2 and downstream pathway signaling in cancer cells, and thus is considered to be effective as a cancer therapeutic agent.
Therefore, there is a need to further develop new EGFR inhibitors and HER2 inhibitors in order to expect that exon 20 insertion mutated EGFR, exon 18 point mutated EGFR, exon 21 point mutated EGFR, wild HER2 and/or mutated HER2 can be effectively inhibited.
Summary of The Invention
The invention provides a novel fused tricyclic derivative, a composition containing the compound and application thereof, wherein the fused tricyclic derivative has better inhibitory activity and high selectivity on exon 20 insertion (exon 20ins) mutant EGFR, exon 18 site mutant EGFR, exon 21 site mutant EGFR, exon 19 deletion (exon 19del) mutant EGFR, L858R mutant EGFR, exon 19 deletion/T790M mutant EGFR and L858R/T790M mutant EGFR, has no inhibitory activity or little inhibitory activity on wild-type EGFR, and has inhibitor activity on wild-type HER2 and/or mutant HER2, so that the anti-tumor inhibitor with low toxic and side effects is provided.
In contrast, the invention adopts the following technical scheme:
in one aspect, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure BDA0002697128120000021
wherein, the first and the second end of the pipe are connected with each other,
ring A is selected from C 6-14 Aryl or 5 to 10 membered heteroaryl;
each R is independently selected from H, D, halogen, -OH, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy or C 1-6 A haloalkoxy group;
p is selected from 0, 1,2, 3, 4 or 5;
R 1 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl, wherein said C is 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more substituents selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
x is C (R) 4 )(R 4 ’);
Y is C (R) 5 )(R 5 ’);
Figure BDA0002697128120000031
Represents a single or double bond;
Figure BDA0002697128120000032
represents a stereoconfiguration of R or S;
when in use
Figure BDA0002697128120000033
When representing a double bond, X is C (R) 4 );
m is 0, 1 or 2;
n is 1,2 or 3;
when m is 0, R 1 The carbon atom to which it is attached directly forms a bond with the carbon atom to which the amide N atom is attached;
R 4 and R 4 ' are each independently selected from H, D, halogen, -CN or C 1-6 An alkyl group; or R 4 And R 4 ' form carbonyl groups, C, with the carbon atom to which they are attached 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 5 and R 5 ' are each independently selected from H, D, halogen, -CN or C 1-6 An alkyl group; or R 5 And R 5 ' form carbonyl groups, C, with the carbon atom to which they are attached 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
provided that when
Figure BDA0002697128120000041
R represents a single bond and m is 1 1 Is not H.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient. In a particular embodiment, the compounds of the present invention are provided in a therapeutically effective amount. In particular embodiments, the compounds of the present invention are provided in a prophylactically effective amount.
In another aspect, the present invention provides a use of a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention, for the preparation of a medicament for the treatment and/or prevention of a mutated EGFR kinase mediated tumor.
In another aspect, the present invention provides a method of treating and/or preventing a disease in a subject, such as a mutant EGFR kinase-mediated tumor, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
In a specific embodiment, the exon 20 insertion mutation is a mutation wherein one or more amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is a mutation wherein 1 to 7 amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is a mutation wherein 1 to 4 amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is A763_ Y764insFQEA, V769_ D770insASV, D770_ N771insSVD, D770_ N771insNPG, D770_ N771insG, D770> GY, N771_ P772insN, P772_ R773insPR, H773_ V774insNPH, H773_ V774insPH, H773_ V774insAH, H773_ V774insH, H774_ C774insHV, A761_ E762 insEAFQ. In specific embodiments, the exon 20 insertion mutation is V769_ D770insASV, D770_ N771insSVD, D770_ N771insG, H773_ V774insNPH, or H773_ V774 insPH.
In a specific embodiment, said exon 18 point mutation is selected from the group consisting of the exon 18G 719X mutation and the exon 18E 790X mutation. In a specific embodiment, the mutation at G719X is at least one mutation selected from the group consisting of G719A, G719S and G719C. In particular embodiments, the E790X mutation is selected from at least one mutation of E790K and E790A.
In a specific embodiment, said exon 21 point mutation is selected from the group consisting of the L861X mutation of exon 21. In a specific embodiment, the L861X mutation is an L861Q mutation.
In specific embodiments, the mutant EGFR has a T790M mutation and has at least one mutation selected from an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation or a L858R mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with an exon 20 insertion mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with the T790M mutation and with an exon 20 insertion mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a patient with a tumor expressing EGFR having an exon 18 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a tumor patient expressing EGFR having the T790M mutation and having an exon 18 point mutation.
In a specific embodiment, a compound of the invention, or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, is used to treat a tumor patient expressing EGFR with an exon 21 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used to treat cancer patients expressing EGFR having the T790M mutation and having an exon 21 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a tumor patient expressing EGFR having an exon 19 deletion mutant.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used to treat cancer patients expressing EGFR having the T790M mutation and having an exon 19 deletion mutant.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with L858R mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with the T790M mutation and with the L858R mutation.
In a specific embodiment, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention for the preparation of a medicament for the treatment and/or prevention of the following tumors, or a method for the treatment and/or prevention of the following tumors in a subject, comprising administering a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention to the subject: lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
In another aspect, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention, for the preparation of a medicament for the treatment and/or prevention of a tumor mediated by wild-type and/or mutant HER2 kinase.
In another aspect, the present invention provides a method of treating and/or preventing a disease in a subject, such as a wild-type and/or mutant HER2 kinase-mediated tumor, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20 insymva mutant HER 2.
In particular embodiments, the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
In a specific embodiment, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention for the preparation of a medicament for the treatment and/or prevention of the following tumors, or a method for the treatment and/or prevention of the following tumors in a subject, comprising administering a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention to the subject: lung cancer, gastric cancer or breast cancer.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Definition of
Chemical definition
The definitions of specific functional groups and chemical terms are described in more detail below.
When a range of values is recited, it is intended to include each value and every subrange within the range. E.g. "C 1-6 Alkyl "includes C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 1-6 、C 1-5 、C 1-4 、C 1-3 、C 1-2 、C 2-6 、C 2-5 、C 2-4 、C 2-3 、C 3-6 、C 3-5 、C 3-4 、C 4-6 、C 4-5 And C 5-6 An alkyl group.
“C 1-6 Alkyl "refers to a straight or branched chain saturated hydrocarbon group having 1 to 6 carbon atoms, also referred to herein as" lower alkyl ". In some embodiments, C 1-4 Alkyl groups are particularly preferred. Examples of such alkyl groups include, but are not limited to: methyl (C) 1 ) Ethyl (C) 2 ) N-propyl (C) 3 ) Isopropyl (C) 3 ) N-butyl (C) 4 ) Tert-butyl (C) 4 ) Sec-butyl (C) 4 ) Isobutyl (C) 4 ) N-pentyl group (C) 5 ) 3-pentyl radical (C) 5 ) Pentyl radical (C) 5 ) Neopentyl (C) 5 ) 3-methyl-2-butyl (C) 5 ) Tert-amyl (C) 5 ) And n-hexyl (C) 6 ). Each of the alkyl groups is independently optionally substituted, whether or not the alkyl group is preceded by a "substituted", e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
“C 1-6 Alkoxy "refers to the group-OR, where R is substituted OR unsubstituted C 1-6 An alkyl group. In some embodiments, C 1-4 Alkoxy groups are particularly preferred. Specific said alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1, 2-dimethylbutoxy.
"halo" or "halogen" refers to fluoro (F), chloro (Cl), bromo (Br), and iodo (I). In some embodiments, the halogen group is F, Cl or Br. In some embodiments, the halogen group is F or Cl. In some embodiments, the halogen group is F.
Thus, "C 1-6 Haloalkyl "and" C 1-6 Haloalkoxy "means" C "as defined above 1-6 Alkyl "and" C 1-6 Alkoxy ", which is substituted with one or more halo groups. In some embodiments, C 1-4 Haloalkyl is particularly preferred, more preferably C 1-2 A haloalkyl group. In some embodiments, C 1-4 Haloalkoxy is particularly preferred, more preferably C 1-2 A haloalkoxy group. Exemplary said haloalkyl groups include, but are not limited to: -CF 3 、-CH 2 F、-CHF 2 、-CHFCH 2 F、-CH 2 CHF 2 、-CF 2 CF 3 、-CCl 3 、-CH 2 Cl、-CHCl 2 2,2, 2-trifluoro-1, 1-dimethyl-ethyl, and the like. Exemplary said haloalkoxy groups include, but are not limited to: -OCH 2 F、-OCHF 2 、-OCF 3 And so on.
“C 3-10 Cycloalkyl "refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-7 Cycloalkyl is preferred, C 3-6 Cycloalkyl is particularly preferred, more preferably C 5-6 A cycloalkyl group. Cycloalkyl also includes ring systems in which the aforementioned cycloalkyl ring is fused to one or more aryl or heteroaryl groups, where the point of attachment is on the cycloalkyl ring, and in such cases the number of carbons continues to represent the number of carbons in the cycloalkyl system. Exemplary such cycloalkyl groups include, but are not limited to: cyclopropyl (C) 3 ) Cyclopropenyl group (C) 3 ) Cyclobutyl (C) 4 ) Cyclobutenyl radical (C) 4 ) Cyclopentyl (C) 5 ) Cyclopentenyl group (C) 5 ) Cyclohexyl (C) 6 ) Cyclohexenyl (C) 6 ) Cyclohexyldienyl (C) 6 ) Cycloheptyl (C) 7 ) Cycloheptenyl (C) 7 ) Cycloheptadienyl (C) 7 ) Cycloheptatrienyl (C) 7 ) Cyclooctyl (C) 8 ) Cyclooctenyl (C) 8 ) Bicyclo [2.2.1]Heptyl (C) 7 ) Bicyclo [2.2.2 ] s]Octyl radical (C) 8 ) Cyclononyl (C) 9 ) Cyclononenyl (C) 9 ) Cyclodecyl (C) 10 ) Cyclodecenyl (C) 10 ) octahydro-1H-indenyl (C) 9 ) Decahydronaphthyl (C) 10 ) Spiro [4.5 ]]Decyl (C) 10 ) And so on. Each of the cycloalkyl groups is independently optionally substituted, whether or not the cycloalkyl group is modified "substituted", for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
"3-to 10-membered heterocyclic" or "heterocyclic" refers to a group having a ring carbon atom and 1 to 4 ring heteroatoms in a 3-to 10-membered non-aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon. In heterocyclic groups containing one or more nitrogen atoms, the point of attachment may be carbon or a nitrogen atom, as valency permits. In some embodiments, 3 to 7 membered heterocyclic groups are preferred, which are 3 to 7 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms; in some embodiments, 3 to 6 membered heterocyclic groups are particularly preferred, which are 3 to 6 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms; more preferably a5 to 6 membered heterocyclic group which is a5 to 6 membered non aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms. Heterocyclyl also includes ring systems wherein the aforementioned heterocyclyl ring is fused to one or more cycloalkyl, aryl, or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring; and in such cases the number of ring members continues to represent the number of ring members in the heterocyclyl ring system. Each of the heterocyclic groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the heterocyclic group is pre-modified with "substituted".
Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to: aziridinyl, oxacyclopropaneyl, thienylyl. Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl and thietanyl. Exemplary containing5-membered heterocyclic groups having one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2, 5-dione. Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to: dioxolanyl, oxathiolanyl (oxathiolanyl), dithiolanyl (disulphuryl), and oxazolidin-2-one. Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl and thiadiazolinyl. Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridinyl and thiacyclohexyl (thianyl). Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithiacyclohexane, dioxanyl. Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to: hexahydrotriazinyl (triazinanyl). Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to: azepane, oxepanyl and thiepane. Exemplary 8-membered heterocyclic groups containing one heteroatom include, but are not limited to: azacyclooctyl, oxocyclooctyl, and thiacyclooctyl. Exemplary with C 6 Aryl ring fused 5-membered heterocyclyl (also referred to herein as 5, 6-bicyclic heterocyclyl) includes, but is not limited to: indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolonyl, and the like. Exemplary with C 6 Aryl ring fused 6-membered heterocyclyl (also referred to herein as6, 6-bicyclic heterocyclyl) includes, but is not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
“C 6-14 Aryl "refers to a group having 6 to 14 ring carbon atoms and zero heteroatoms of a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n +2 aromatic ring system (e.g., having 6, 10, or 14 pi electrons shared in a cyclic arrangement). In some embodiments, an aryl group has six ring carbon atoms ("C) 6 Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C) 10 Aryl "; e.g., naphthyl, e.g., 1-naphthyl and 2-naphthyl). In thatIn some embodiments, aryl has fourteen ring carbon atoms ("C) 14 Aryl "; for example, an anthracene group). In some embodiments, C 6-10 Aryl is particularly preferred, more preferably C 6 And (3) an aryl group. Aryl also includes ring systems in which the aforementioned aryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system. Each of the aryl groups is independently optionally substituted, whether or not the aryl group is modified "substituted", for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
"5-to 10-membered heteroaryl" refers to a group having a 5-10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) with ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur. In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems may include one or more heteroatoms in one or both rings. Heteroaryl also includes ring systems in which the aforementioned heteroaryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the heteroaryl ring system. In some embodiments, 5-to 6-membered heteroaryl groups are particularly preferred, which are 5-6 membered monocyclic or bicyclic 4n +2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms. Each of the heteroaryl groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the heteroaryl group is modified "substituted" before the heteroaryl group.
Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furanyl and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: a tetrazolyl group. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: a pyridyl group. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to: triazinyl and tetrazinyl. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxacycloheptyl, and thiacycloheptyl trienyl groups. Exemplary 5, 6-bicyclic heteroaryls include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzisothiafuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, indezinyl, and purinyl. Exemplary 6, 6-bicyclic heteroaryls include, but are not limited to: naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl and quinazolinyl.
"carbonyl" means a-C (O) -group.
Exemplary substituents on carbon atoms include, but are not limited to: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR aa 、-ON(R bb ) 2 、-N(R bb ) 2 、-N(R bb ) 3 + X - 、-N(OR cc )R bb 、-SH、-SR aa 、-SSR cc 、-C(=O)R aa 、-CO 2 H、-CHO、-C(OR cc ) 2 、-CO 2 R aa 、-OC(=O)R aa 、-OCO 2 R aa 、-C(=O)N(R bb ) 2 、-OC(=O)N(R bb ) 2 、-NR bb C(=O)R aa 、-NR bb CO 2 R aa 、-NR bb C(=O)N(R bb ) 2 、-C(=NR bb )R aa 、-C(=NR bb )OR aa 、-OC(=NR bb )R aa 、-OC(=NR bb )OR aa 、-C(=NR bb )N(R bb ) 2 、-OC(=NR bb )N(R bb ) 2 、-NR bb C(=NR bb )N(R bb ) 2 、-C(=O)NR bb SO 2 R aa 、-NR bb SO 2 R aa 、-SO 2 N(R bb ) 2 、-SO 2 R aa 、-SO 2 OR aa 、-OSO 2 R aa 、-S(=O)R aa 、-OS(=O)R aa 、-Si(R aa ) 3 、-OSi(R aa ) 3 、-C(=S)N(R bb ) 2 、-C(=O)SR aa 、-C(=S)SR aa 、-SC(=S)SR aa 、-SC(=O)SR aa 、-OC(=O)SR aa 、-SC(=O)OR aa 、-SC(=O)R aa 、-P(=O) 2 R aa 、-OP(=O) 2 R aa 、-P(=O)(R aa ) 2 、-OP(=O)(R aa ) 2 、-OP(=O)(OR cc ) 2 、-P(=O) 2 N(R bb ) 2 、-OP(=O) 2 N(R bb ) 2 、-P(=O)(NR bb ) 2 、-OP(=O)(NR bb ) 2 、-NR bb P(=O)(OR cc ) 2 、-NR bb P(=O)(NR bb ) 2 、-P(R cc ) 2 、-P(R cc ) 3 、-OP(R cc ) 2 、-OP(R cc ) 3 、-B(R aa ) 2 、-B(OR cc ) 2 、-BR aa (OR cc ) Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3, 4 or 5R dd Substituted by groups;
or two geminal hydrogens on carbon atoms are selected from the group consisting of O, S, NN (R) bb ) 2 、=NNR bb C(=O)R aa 、=NNR bb C(=O)OR aa 、=NNR bb S(=O) 2 R aa 、=NR bb Or as NOR cc Substitution;
R aa each independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R aa The groups combine to form a heterocyclyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2, 3, 4, or 5R dd Substituted by groups;
R bb each independently selected from: hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R bb The groups combine to form a heterocyclyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2, 3, 4, or 5R dd Substituted by groups;
R cc each is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R cc The groups combine to form a heterocyclyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2, 3, 4, or 5R dd Substituted by groups;
R dd each independently selected from: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR ee 、-ON(R ff ) 2 、-N(R ff ) 2 ,、-N(R ff ) 3 + X - 、-N(OR ee )R ff 、-SH、-SR ee 、-SSR ee 、-C(=O)R ee 、-CO 2 H、-CO 2 R ee 、-OC(=O)R ee 、-OCO 2 R ee 、-C(=O)N(R ff ) 2 、-OC(=O)N(R ff ) 2 、-NR ff C(=O)R ee 、-NR ff CO 2 R ee 、-NR ff C(=O)N(R ff ) 2 、-C(=NR ff )OR ee 、-OC(=NR ff )R ee 、-OC(=NR ff )OR ee 、-C(=NR ff )N(R ff ) 2 、-OC(=NR ff )N(R ff ) 2 、-NR ff C(=NR ff )N(R ff ) 2 、-NR ff SO 2 R ee 、-SO 2 N(R ff ) 2 、-SO 2 R ee 、-SO 2 OR ee 、-OSO 2 R ee 、-S(=O)R ee 、-Si(R ee ) 3 、-OSi(R ee ) 3 、-C(=S)N(R ff ) 2 、-C(=O)SR ee 、-C(=S)SR ee 、-SC(=S)SR ee 、-P(=O) 2 R ee 、-P(=O)(R ee ) 2 、-OP(=O)(R ee ) 2 、-OP(=O)(OR ee ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3, 4 or 5R gg Substituted by radicals, or two geminal R dd Substituents may combine to form ═ O or ═ S;
R ee is independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, heterocyclyl, carbocyclyl, aryl, and heteroarylBy 0, 1,2, 3, 4 or 5R gg Substitution of radicals;
R ff each is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R ff The groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3, 4 or 5R gg Substituted by groups;
R gg is independently of each other: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OC 1-6 Alkyl, -ON (C) 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 3 + X - 、-NH(C 1-6 Alkyl radical) 2 + X - 、-NH 2 (C 1-6 Alkyl radical) + X - 、-NH 3 + X - 、-N(OC 1-6 Alkyl) (C 1-6 Alkyl), -N (OH) (C) 1-6 Alkyl), -NH (OH), -SH, -SC 1-6 Alkyl, -SS (C) 1-6 Alkyl), -C (═ O) (C) 1-6 Alkyl), -CO 2 H、-CO 2 (C 1-6 Alkyl), -OC (═ O) (C) 1-6 Alkyl), -OCO 2 (C 1-6 Alkyl), -C (═ O) NH 2 、-C(=O)N(C 1-6 Alkyl radical) 2 、-OC(=O)NH(C 1-6 Alkyl), -NHC (═ O) (C) 1-6 Alkyl), -N (C) 1-6 Alkyl) C (═ O) (C) 1-6 Alkyl), -NHCO 2 (C 1-6 Alkyl), -NHC (═ O) N (C) 1-6 Alkyl radical) 2 、-NHC(=O)NH(C 1-6 Alkyl), -NHC (═ O) NH 2 、-C(=NH)O(C 1-6 Alkyl), -OC (═ NH) (C) 1-6 Alkyl), -OC (═ NH) OC 1-6 Alkyl, -C (═ NH) N (C) 1-6 Alkyl radical) 2 、-C(=NH)NH(C 1-6 Alkyl), -C (═ NH) NH 2 、-OC(=NH)N(C 1-6 Alkyl radical) 2 、-OC(NH)NH(C 1-6 Alkyl), -OC (NH) NH 2 、-NHC(NH)N(C 1-6 Alkyl radical) 2 、-NHC(=NH)NH 2 、-NHSO 2 (C 1-6 Alkyl), -SO 2 N(C 1-6 Alkyl radical) 2 、-SO 2 NH(C 1-6 Alkyl), -SO 2 NH 2 、-SO 2 C 1-6 Alkyl, -SO 2 OC 1-6 Alkyl, -OSO 2 C 1-6 Alkyl, -SOC 1-6 Alkyl, -Si (C) 1-6 Alkyl radical) 3 、-OSi(C 1-6 Alkyl radical) 3 、-C(=S)N(C 1-6 Alkyl radical) 2 、C(=S)NH(C 1-6 Alkyl), C (═ S) NH 2 、-C(=O)S(C 1-6 Alkyl), -C (═ S) SC 1-6 Alkyl, -SC (═ S) SC 1-6 Alkyl, -P (═ O) 2 (C 1-6 Alkyl), -P (═ O) (C) 1-6 Alkyl radical) 2 、-OP(=O)(C 1-6 Alkyl radical) 2 、-OP(=O)(OC 1-6 Alkyl radical) 2 、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2 -C 6 Alkenyl radical, C 2 -C 6 Alkynyl, C 3 -C 7 Carbocyclic radical, C 6 -C 10 Aryl radical, C 3 -C 7 Heterocyclic group, C 5 -C 10 A heteroaryl group; or two geminal R gg Substituents may combine to form ═ O or ═ S; wherein, X - Are counter ions.
Exemplary substituents on the nitrogen atom include, but are not limited to: hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR bb )R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, cycloalkyl, heterocycloalkyl, heterocyclo, or a pharmaceutically acceptable salt thereof,Heterocyclyl, aryl and heteroaryl, or two R attached to a nitrogen atom cc The groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3, 4 or 5R dd Is substituted by radicals, and wherein R aa 、R bb 、R cc And R dd As described above.
"deuterated" or "D" means that one or more hydrogens of the compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
"non-deuterated compound" refers to a compound containing deuterium at an atomic ratio not higher than the natural isotopic content of deuterium (0.015%).
The deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
The term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977)66: 1-19. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Salts formed using methods conventional in the art, for example, ion exchange methods, are also included. Other pharmaceutically acceptable salts include: adipate, ascorbate, aspartate, benzenesulfonate, benzoate, and bisulfateBorate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentylpropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, gluconate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, ditolyl-valerate, and mixtures thereof, Valeric acid salts, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl radical) 4 And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like. Other pharmaceutically acceptable salts include, if appropriate, non-toxic ammonium, quaternary ammonium and amine cations formed with counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
The "subject" to which the drug is administered includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms "human", "patient" and "subject" are used interchangeably herein.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes the effect that occurs when a subject has a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
"combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously with another therapeutic agent in a single unit dosage form.
Detailed Description
Compound (I)
As used herein, "compounds of the invention" refers to compounds of formula (I) below (including a subset of each formula), or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
In one embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure BDA0002697128120000141
wherein the content of the first and second substances,
ring A is selected from C 6-14 Aryl or 5 to 10 membered heteroaryl;
each R is independently selected from H, D, halogen, -OH, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy or C 1-6 A haloalkoxy group;
p is selected from 0, 1,2, 3, 4 or 5;
R 1 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH、-NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
x is C (R) 4 )(R 4 ’);
Y is C (R) 5 )(R 5 ’);
Figure BDA0002697128120000151
Represents a single bond or a double bond;
Figure BDA0002697128120000152
represents the stereoconfiguration of R or S;
when in use
Figure BDA0002697128120000153
When representing a double bond, X is C (R) 4 );
m is 0, 1 or 2;
n is 1,2 or 3;
when m is 0, R 1 The carbon atom to which it is attached directly forms a bond with the carbon atom to which the amide N atom is attached;
R 4 and R 4 ' are each independently selected from H, D, halogen, -CN or C 1-6 An alkyl group; or R 4 And R 4 ' form carbonyl groups, C, with the carbon atom to which they are attached 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more substituents selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 5 and R 5 ' are each independently selected from H, D, halogen, -CN or C 1-6 An alkyl group; or R 5 And R 5 ' form carbonyl groups, C, with the carbon atom to which they are attached 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached; wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
provided that when
Figure BDA0002697128120000161
R represents a single bond and m is 1 1 Is not H.
Ring A
In one embodiment, ring A is selected from C 6-14 Aryl or 5 to 10 membered heteroaryl; in another embodiment, ring A is C 6-14 An aryl group; in another embodiment, ring a is a5 to 10 membered heteroaryl; in another embodiment, ring a is selected from phenyl, naphthyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyrrolyl, pyrazolyl, imidazolyl, furanyl, oxazolyl, isoxazolyl, thienyl, thiazolyl, isothiazolyl, quinolinyl, or isoquinolinyl; in another embodiment, ring a is selected from phenyl, naphthyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, quinolinyl, or isoquinolinyl; in another embodiment, ring a is selected from naphthyl, quinolinyl, or isoquinolinyl; in another embodiment, ring a is selected from quinolinyl or isoquinolinyl; in another embodiment, ring a is quinolinyl.
R
In one embodiment, each R is independently selected from H, D, halogen, -OH, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy or C 1-6 A haloalkoxy group; in another embodiment, each R is independently H; in another embodiment, each R is independently D; in another embodiment, each R is independently halogen; in another embodiment, each R is independently-OH; in another embodiment, each R is independently-CN; in another embodiment, each R is independently C 1-6 An alkyl group; in another embodiment, each R is independently C 1-6 A haloalkyl group; in another embodiment, each R is independently C 1-6 An alkoxy group; in another embodiment, each R is independently C 1-6 A haloalkoxy group.
In another embodiment, each R is independently selected from D, halo, -OH, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy or C 1-6 HaloalkoxyA group; in another embodiment, each R is independently selected from D, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy or C 1-6 A haloalkoxy group; in another embodiment, each R is independently selected from D, halogen, C 1-6 Alkyl or C 1-6 A haloalkyl group; in another embodiment, each R is independently selected from D, halogen or C 1-6 An alkyl group; in another embodiment, each R is independently selected from D, F, Cl, Br, -CH 3 、-CF 3 、-CHF 2 、-CH 2 F、-OCH 3 、-CH(CH 3 ) 2 or-OCH (CH) 3 ) 2 (ii) a In another embodiment, each R is independently selected from the group consisting of D, F, -CH 3 、-CF 3 、-OCH 3 or-CH (CH) 3 ) 2 (ii) a In another embodiment, each R is independently selected from D, F or-CH 3
p
In one embodiment, p is selected from 0, 1,2, 3, 4 or 5; in another embodiment, p is 0; in another embodiment, p is 1; in another embodiment, p is 2; in another embodiment, p is 3; in another embodiment, p is 4; in another embodiment, p is 5.
R 1
In one embodiment, R 1 Selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Is H; in another embodiment, R 1 Is D; in another embodiment, R 1 Is halogen; in another embodiment, R 1 is-CN; in another embodiment, R 1 Is C 1-6 An alkyl group; in another embodiment, R 1 Is C 1-6 An alkoxy group; in another embodiment, R 1 Is C 3-7 A cycloalkyl group; in another embodiment, R 1 Is a 3 to 7 membered heterocyclyl; in another embodiment, R 1 C in (1) 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 Alkoxy groups.
In another embodiment, R 1 Selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more substituents selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Selected from H, D, halogen, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more substituents selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Selected from H, D or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Selected from H, D or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH or-NR a R b Substituted with a group of (1); in another embodiment, R 1 Selected from H, D or C 1-6 An alkyl group; in another embodiment, R 1 Selected from H, D, methyl, ethyl or isopropyl; in another embodiment, R 1 Selected from H, D or methyl; in another embodiment, R 1 Selected from H or methyl.
In another embodiment, R 1 Selected from halogen, -CN, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl, wherein said C is 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Is selected from C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Is selected from C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 1 Is C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b Substituted with a group of (1); in another embodiment, R 1 Is C 1-6 An alkyl group; in another embodiment, R 1 Selected from methyl, ethyl or isopropyl; in another embodiment, R 1 Is methyl.
R 2
In one embodiment, R 2 Selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl are each independently optionally substituted with one or more substituents selected from D, halogen,-OH、-NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond; in another embodiment, R 2 Is H; in another embodiment, R 2 Is D; in another embodiment, R 2 Is halogen; in another embodiment, R 2 is-CN; in another embodiment, R 2 Is C 1-6 An alkyl group; in another embodiment, R 2 Is C 3-7 A cycloalkyl group; in another embodiment, R 2 Is a 3 to 7 membered heterocyclyl; in another embodiment, R 2 Is C 6-10 An aryl group; in another embodiment, R 2 Is a5 to 10 membered heteroaryl; in another embodiment, R 2 C in (1) 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 2 Together with the double bond to which it is attached, form a triple bond.
In another embodiment, R 2 Independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl are each independently optionally substituted with one or more substituents selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 2 Independently selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 2 Independently selected from H, D, halogen or C 1-6 Alkyl radical, whereinC is 1-6 Alkyl is optionally substituted by one or more groups selected from D, halogen, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 2 Independently selected from H, D, halogen or C 1-6 An alkyl group; in another embodiment, R 2 Independently selected from H, D, F, Cl or methyl.
R 3
In one embodiment, R 3 Selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 3 Is H; in another embodiment, R 3 Is D; in another embodiment, R 3 Is halogen; in another embodiment, R 3 is-CN; in another embodiment, R 3 Is C 1-6 An alkyl group; in another embodiment, R 3 Is C 3-7 A cycloalkyl group; in another embodiment, R 3 Is a 3 to 7 membered heterocyclyl; in another embodiment, R 3 Is C 6-10 An aryl group; in another embodiment, R 3 Is a5 to 10 membered heteroaryl; in another embodiment, R 3 C in (1) 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 Alkoxy groups.
In another embodiment, R 3 Selected from H, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 3 Selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R 3 Selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from D, halogen, -NR a R b Or C 1-6 Radical substitution of alkyl; in another embodiment, R 3 Selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, wherein said C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl are each independently optionally substituted by one or more groups selected from-NR a R b Substituted by groups; in another embodiment, R 3 Is selected from H or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from-NR a R b Substituted by groups; in another embodiment, R 3 Selected from H, methyl, ethyl, -CH 2 N(CH 3 ) 2 、-CH 2 N(CH 3 )(CH 2 CH 3 )、-CH 2 N(CH 3 )(CH(CH 3 ) 2 )、-CH 2 N(CH 2 CH 3 ) 2 Or
Figure BDA0002697128120000201
In another embodiment, R 3 Is H.
R’
In one embodiment, R' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; in another embodiment, R' is H; in another embodiment, R' is C 1-6 An alkyl group; in another embodiment, R' is C 1-6 A haloalkyl group; in another embodiment, C in R' is 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 Alkoxy groups.
In another embodiment, R' is selected from H or C 1-6 An alkyl group; in another embodiment, R' is selected from H, methyl, ethyl or isopropyl; in another embodiment, R' is selected from H or methyl.
m
In one embodiment, m is 0, 1 or 2; in another embodiment, m is 0; in another embodiment, m is 1; in another embodiment, m is 2.
n
In one embodiment, n is 1,2 or 3; in another embodiment, n is 1; in another embodiment, n is 2; in another embodiment, n is 3.
Any of the above embodiments, or any combination thereof, may be combined with any of the other embodiments, or any combination thereof. For example, any embodiment of ring A, or any combination thereof, may be combined with R, R 1 To R 3 R', X, Y, m, n and p or a pharmaceutically acceptable salt thereofAny combination is combined. The present invention is intended to include all combinations of these solutions, limited to space, not listed one by one.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000211
wherein each group is as defined above.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000212
wherein each group is as defined above;
preferably, the first and second electrodes are formed of a metal,
R 1 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
preferably, the first and second liquid crystal display panels are,
R 1 is C 1-6 Alkyl, preferably methyl;
r' is H;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000221
wherein each group is as defined above;
preferably, the first and second liquid crystal display panels are,
R 1 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl andC 1-6 haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; preferably, R' is selected from C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
preferably, the first and second electrodes are formed of a metal,
R 1 is C 1-6 Alkyl, preferably methyl;
r' is H; or R' is selected from C 1-6 Alkyl or C 1-6 A haloalkyl group;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000241
wherein each group is as defined above.
In a more specific embodiment, the present invention relates to a compound described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000242
wherein each group is as defined above;
preferably, the first and second electrodes are formed of a metal,
R 1 selected from D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; preferably, R 1 Is in S configuration; preferably, R 1 Is in the R configuration;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
preferably, the first and second liquid crystal display panels are,
R 1 is C 1-6 Alkyl, preferably methyl; preferably, R 1 Is in S configuration; preferably, R 1 Is in the R configuration;
r' is H;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000251
wherein each group is as defined above;
preferably, the first and second electrodes are formed of a metal,
R 1 selected from D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; preferably, R' is selected from C 1-6 Alkyl or C 1-6 A halogenated alkyl group,wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which it is attached, form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
preferably, the first and second liquid crystal display panels are,
R 1 is C 1-6 Alkyl, preferably methyl;
r' is H; or R' is selected from C 1-6 Alkyl or C 1-6 A haloalkyl group;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000261
wherein each group is as defined above.
In a more specific embodiment, the present invention relates to a compound as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000271
wherein each group is as defined above;
preferably, the first and second electrodes are formed of a metal,
R 1 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl, wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
preferably, the first and second electrodes are formed of a metal,
R 1 is C 1-6 Alkyl, preferably methyl;
r' is H;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002697128120000281
wherein each group is as defined above;
preferably, the first and second electrodes are formed of a metal,
R 1 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted by one or more groups selected from D, halo, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
r' is selected from H, C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; preferably, R' is selected from C 1-6 Alkyl or C 1-6 Haloalkyl, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl is each independently optionally substituted with one or more groups selected from D, halogen, C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R 2 selected from H, D, halogen or C 1-6 Alkyl, wherein said C 1-6 Alkyl is independently optionally substituted by one or more groups selected from D, haloElement, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy; or R 2 Together with the double bond to which they are attached form a triple bond;
R 3 selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl wherein said C is 1-6 Alkyl radical, C 3-7 Cycloalkyl and 3-to 7-membered heterocyclyl are each independently optionally substituted with one or more groups selected from D, halogen, -OH, -NR a R b 、C 1-6 Alkyl or C 1-6 A radical substitution of alkoxy;
R a and R b Each independently selected from H, C 1-6 Alkyl radical, C 3-7 Cycloalkyl or 3 to 7 membered heterocyclyl; or R a 、R b Form a 3-to 7-membered heterocyclic group with the nitrogen atom to which they are attached;
provided that R is 1 Is not H;
preferably, the first and second electrodes are formed of a metal,
R 1 is C 1-6 Alkyl, preferably methyl;
r' is H; or R' is selected from C 1-6 Alkyl or C 1-6 A haloalkyl group;
R 2 is H;
R 3 is H.
In a more specific embodiment, the present invention relates to a compound selected from the group consisting of:
Figure BDA0002697128120000291
the compounds of the invention may include one or more asymmetric centers and may therefore exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be in the form of individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
"tautomer" refers to a compound in which one functional group changes its structure to another functional isomer, and which rapidly interconverts into two isomers in dynamic equilibrium, the two isomers being referred to as tautomers.
One skilled in the art will appreciate that the organic compound may form a complex with a solvent in which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is combined with a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates.
The term "hydrate" refers to a compound that is associated with an aqueous phase. In general, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, hydrates of the compounds can be used, for example, of the formula R.times. x H 2 O represents, wherein R is the compound, and x isA number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrates (x is a number greater than 0 and less than 1), e.g., hemihydrate (R × 0.5H) 2 O)) and polyhydrates (x is a number greater than 1, e.g. dihydrate (R.times.2H) 2 O) and hexahydrate (Rx6H) 2 O))。
The compounds of the present invention may be in amorphous or crystalline form (crystalline or polymorphic). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may cause a crystalline form to dominate. Various polymorphs of a compound may be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those recited in formula (I), but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H、 3 H、 13 C、 11 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and 36 and (4) Cl. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the invention, e.g. by incorporation of radioactive isotopes (e.g. by introducing 3 H and 14 C) can be used for drug and/or substrate tissue distribution assaysAnd (4) determining. Tritium, i.e. 3 H and carbon-14, i.e. 14 The C isotopes are particularly preferred because of their ease of preparation and detection. Further, by heavier isotopes, e.g. deuterium, i.e. 2 H, may be preferred in some cases because of the higher metabolic stability that may provide therapeutic benefits, such as increased in vivo half-life or reduced dosage requirements. Isotopically-labelled compounds of formula (I) of the present invention and prodrugs thereof can generally be prepared by substituting a readily available isotopically-labelled reagent for a non-isotopically-labelled reagent in the course of performing the procedures disclosed in the schemes and/or in the examples and preparations below.
In addition, prodrugs are also included within the context of the present invention. The term "prodrug" as used herein refers to a compound that is converted in vivo by hydrolysis, for example in the blood, to its active form with a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, Prodrugs as Novel Delivery Systems, vol.14 of a.c.s.symposium Series, Edward b.roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and d.fleisher, s.ramon and h.bara "Improved oral Delivery: the solubility limits of the above overcom by the use of the drugs ", Advanced Drug Delivery Reviews (1996)19, (2)115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded compound of the present invention that releases the parent compound in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner such that the modification is effected by routine manipulation or in vivo cleavage to produce the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form a hydroxy, amino, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of hydroxy, mercapto, and amino functional groups of the compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may be used. The ester itself may be active and/or may be hydrolysed under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which readily break down in the human body to release the parent acid or salt thereof.
Treatment of
The present invention provides a method of treating and/or preventing a disease, such as a mutant EGFR kinase-mediated cancer, in a subject, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant EGFR is selected from the group consisting of an exon 20 insertion mutant EGFR, an exon 18 point mutant EGFR, an exon 21 point mutant EGFR, an exon 19 deletion mutant EGFR, or an L858R mutant EGFR.
In particular embodiments, the mutant EGFR has a T790M mutation and has at least one mutation selected from an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation, or an L858R mutation.
As used herein, "EGFR" refers to the human epidermal growth factor receptor protein and is referred to as ErbB-1 or HER 1.
Herein, "wild-type EGFR" refers to EGFR without somatic mutations.
Herein, "exon 20 insertion mutation" refers to a mutation in which one or more amino acids (preferably 1 to 7, more preferably 1 to 4) are inserted into an exon 20 region (e.g., amino acid sequence at position 761 to 823) of EGFR; preferably, the mutation is a mutation in which the amino acid sequence FQEA (in the order of phenylalanine, glutamine, glutamic acid and alanine from the N-terminus) is inserted between alanine at position 763 and tyrosine at position 764in the region of exon 20 (a763_ Y764 insFQEA); preferably, the mutation is a mutation in which the amino acid sequence ASV (in the order of alanine, serine and valine from the N-terminus) is inserted between valine at position 769 and aspartic acid at position 770in the exon 20 region (V769_ D770 insASV); preferably, the mutation is a mutation in which the amino acid sequence SVD (in the order of serine, valine and aspartic acid from the N-terminus) is inserted between asparagine at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insSVD); preferably, the mutation is a mutation in which the amino acid sequence NPG (asparagine, proline and glycine in this order from the N-terminus) is inserted between aspartic acid at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insNPG); preferably, the mutation is a mutation in which amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771 (D770 — N771 insG); preferably, the mutation is a mutation wherein aspartic acid at position 770in the exon 20 region is deleted and thereby the amino acid sequence GY (glycine and tyrosine in this order from the N-terminus) is inserted (D770> GY); preferably, the mutation is a mutation in which amino acid N (asparagine) is inserted between asparagine at position 771 and proline at position 772in the exon 20 region (N771_ P772 insN); preferably, the mutation is a mutation in which the amino acid sequence PR (proline and arginine in this order from the N-terminus) is inserted between proline at position 772 and histidine at position 773in the region of exon 20 (P772_ R773 insPR); preferably, the mutation is a mutation in which an amino acid sequence NPH (asparagine, proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773_ V774 insNPH); preferably, the mutation is a mutation in which the amino acid sequence PH (proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773_ V774 insPH); preferably, the mutation is a mutation in which the amino acid sequence AH (alanine and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773_ V774 insAH); preferably, the mutation is a mutation in which an amino acid H (histidine) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773_ V774 insH); preferably, the mutation is a mutation in which the amino acid sequence HV (in the order of histidine and valine from the N-terminus) is inserted between valine at position 774 and cysteine at position 775 in the exon 20 region (V774_ C774 insHV); preferably, the mutation is a mutation in which the amino acid sequence EAFQ (in the order of glutamic acid, alanine, phenylalanine and glutamine from the N-terminus) is inserted between alanine at position 761 and glutamic acid at position 762in the exon 20 region (A761_ E762 insEAFQ). More preferably, the mutation is a mutation in which the amino acid sequence ASV (in the order of alanine, serine and valine from the N-terminus) is inserted between valine at position 769 and aspartic acid at position 770in the exon 20 region (V769_ D770 insASV); more preferably, the mutation is a mutation in which the amino acid sequence SVD (in the order of serine, valine and aspartic acid from the N-terminus) is inserted between asparagine at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insSVD); more preferably, the mutation is a mutation in which an amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771in the region of exon 20 (D770-N771 insG); more preferably, the mutation is a mutation in which the amino acid sequence NPH (asparagine, proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773V 774 insNPH); more preferably, the mutation is a mutation in which the amino acid sequence PH (proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773_ V774 insPH); more preferably, the mutation is a mutation in which the amino acid sequence SVD (in the order of serine, valine and aspartic acid from the N-terminus) is inserted between aspartic acid at position 770 and aspartic acid at position 771in the exon 20 region (D770-N771 insSVD); more preferably, the mutation is a mutation in which amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insG).
Herein, "cancer patient expressing EGFR with exon 20 insertion mutation" refers to a cancer patient expressing EGFR with exon 20 insertion mutation in at least a part of exon 20 region of EGFR. EGFR may have exon 20 insertion mutations in two or more different portions, but preferably one of them. Moreover, EGFR may also have mutations other than exon 20 insertion mutations (e.g., exon 19 deletion mutation, L858R mutation, or T790M mutation).
In the present invention, the method for detecting an insertion mutation expressing EGFR exon 20in a cancer patient is not particularly limited as long as the method can detect the mutation, and any known detection method can be used. The detection target for detecting the exon 20 insertion mutation may be any one of the gene sequence of the EGFR gene, the transcription product of the EGFR gene, and the EGFR protein.
The sample for detecting the exon 20 insertion mutation is not particularly limited as long as the sample is a biological sample isolated from a cancer patient, particularly a sample obtained from a cancer patient and containing malignant tumor cells. Examples of biological samples include body fluids (e.g., blood, urine, etc.), tissues, extracts thereof, and cultures from which tissues are obtained. The method of isolating the biological sample may be appropriately selected depending on the type of the biological sample.
The biological sample is prepared by appropriate treatment according to the detection method. In addition, a reagent for detection (for example, a reagent containing a primer or a probe) can be prepared by a conventional method according to the detection method.
In one embodiment of the invention, the step of detecting the presence of an exon 20 insertion mutation of EGFR expressed in a patient with a malignant tumor may be performed prior to administering an anti-tumor agent to the cancer patient.
Herein, "exon 18 point mutation" means a point mutation in an amino acid in the exon 18 region of wild-type EGFR. Preferably, the mutation is a point mutation or a deletion mutation in which 1 amino acid in the exon 18 region is substituted; more preferably, the mutation is a point mutation wherein the histidine encoded by codon 709 in exon 18 has been substituted with an arbitrary amino acid (i.e., E790X), and a point mutation wherein the glutamic acid encoded by codon 719 in exon 18 has been substituted with an arbitrary amino acid (i.e., G719X). Specifically, E790X may, for example: a point mutation wherein the glutamic acid encoded by codon 709 in the region of exon 18 has been substituted with lysine (i.e., E709K), and a point mutation wherein the glutamic acid encoded by codon 709 in the region of exon 18 has been substituted with alanine (i.e., E709A). G719X may for example: a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 has been substituted with alanine (i.e., G719A), a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 has been substituted with serine (i.e., G719S), and a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 has been substituted with cysteine (i.e., G719C), with G719A being the most common.
Herein, "exon 21" refers to the region of 824-875 in the amino acid sequence of wild-type EGFR.
Herein, "exon 21 point mutation" means a point mutation in the amino acids of the exon 21 region of wild-type EGFR. Preferably, the exon 21 point mutation is a point mutation wherein 1 amino acid in the exon 21 region is replaced; more preferably, the exon 21 point mutation is a point mutation wherein the leucine encoded by codon 861 in the region of exon 21 has been substituted with any amino acid (i.e.L861X), for example a point mutation wherein the leucine encoded by codon 861 in the region of exon 21 has been substituted with glutamine (i.e.L861Q).
Herein, "exon 18 point mutant EGFR" means EGFR having at least 1 exon 18 point mutation; preferably the EGFR represents a polypeptide having more than 2 related exon 18 point mutations; more preferably, the EGFR indicates a 1 exon 18 point mutation. Furthermore, the EGFR may have other mutations (e.g., exon 19 deletion mutation, L858R mutation, T790M mutation, etc.) other than the exon 18 point mutation.
Herein, "exon 21 point mutant EGFR" means EGFR having at least 1 exon 21 point mutation; preferably the EGFR represents a mutant with more than 2 related exon 21 point mutations; more preferably, the EGFR indicates a 1 exon 21 point mutation. Furthermore, the EGFR may have other mutations (e.g., exon 19 deletion mutation, L858R mutation, T790M mutation, etc.) other than the exon 21 point mutation.
In a specific embodiment, the mutant EGFR is a mutant EGFR having a T790M mutation and having at least one mutation selected from an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation or a L858R mutation.
Specifically, the mutant EGFR having the T790M mutation and having a point selected from the group consisting of exon 18 point mutant EGFR and exon 21 point mutant EGFR of the present invention is any one of the following: mutant EGFR having the T790M mutation and having exon 18 region E709X and/or G719X; mutant EGFR with the T790M mutation and with exon 21 region L861X. Specifically any of the following: mutant EGFR having the T790M mutation and having E709K or E709A; EGFR with the T790M mutation and with G719A, G719S, or G719C mutation; (ii) EGFR with the T790M mutation and with L861Q mutation; among them, mutant EGFR having the T790M mutation and having G719A and having T790M mutation and having L861Q is more common.
In this context, the EGFR expressed by a cancer patient is detected as having a point mutation of exon 18 and/or exon 21 as long as the mutation can be detected, and a known detection method can be used.
The sample for detecting the exon 18 and/or exon 21 point mutation is not particularly limited as long as the sample is a biological sample isolated from a cancer patient, particularly a sample obtained from a cancer patient and containing malignant tumor cells. Examples of biological samples include body fluids (e.g., blood, urine, etc.), tissues, extracts thereof, and cultures from which tissues are obtained. The method of isolating the biological sample may be appropriately selected depending on the type of the biological sample.
The biological sample is prepared by appropriate treatment according to the detection method. In addition, a reagent for detection (for example, a reagent containing a primer or a probe) can be prepared by a conventional method according to the detection method.
In one embodiment of the invention, the step of detecting the presence of exon 18 and/or exon 21 point mutations expressed in a patient with a malignant tumor may be performed prior to administering an anti-neoplastic agent to the cancer patient.
Specific examples of mutated EGFR kinase-mediated tumors of the present invention include, but are not limited to: head and neck cancer, gastrointestinal cancer (esophageal cancer, gastric cancer, duodenal cancer, liver cancer, bile duct cancer (e.g., gallbladder and bile duct cancer), pancreatic cancer, colorectal cancer (e.g., colon cancer and rectal cancer), and the like), lung cancer (e.g., non-small cell lung cancer, and mesothelioma), breast cancer, genital cancer (ovarian cancer, uterine cancer (e.g., cervical cancer, and endometrial cancer), and the like), urinary tract cancer (e.g., renal cancer, bladder cancer, prostate cancer, and testicular cancer), hematopoietic tumors (e.g., leukemia, malignant lymphoma, and multiple myeloma), osteosarcoma, soft tissue sarcoma, skin cancer, brain tumor, and the like. Preferred examples include lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic tumor or skin cancer.
In specific embodiments, the mutant EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
In specific embodiments, the mutant EGFR has the T790M mutation and is selected from the group consisting of an exon 18 point mutant EGFR, an exon 21 point mutant EGFR, an exon 19 deletion mutant EGFR, or a L858R mutant EGFR.
The present invention also provides a method for treating a patient having a tumor comprising the step of administering an effective amount of an antitumor agent comprising a compound of the present invention or a pharmaceutically acceptable salt thereof to a patient expressing a tumor having an exon 20 insertion mutant EGFR, an exon 18 point mutant EGFR, an exon 21 point mutant EGFR, an exon 19 deletion mutant EGFR or an L858R mutant EGFR.
The invention also provides a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in treating a patient expressing a tumor having an expression selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
The invention also provides the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for the treatment of a patient having a tumor selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR or L858R mutant EGFR.
The present invention also provides a method for predicting the effect of treatment with an antitumor agent, which is the compound of the present invention or a pharmaceutically acceptable salt thereof as an active ingredient, in a tumor patient, the method comprising the following steps (1) and (2):
(1) a step of detecting the presence or absence of a mutation of the EGFR gene contained in a biological sample obtained from the patient; and
(2) a step of predicting that chemotherapy is most likely to exhibit a sufficient therapeutic effect on a patient when the result of the detection in step (1) reveals that the EGFR gene has a mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation and an L858R mutation.
The present invention also provides a method for treating a patient having a tumor, the method comprising the following steps (1) to (2):
(1) a step of detecting the presence or absence of a mutation of the EGFR gene contained in a biological sample obtained from the patient;
(2) a step of treating the patient with the compound of the present invention or a pharmaceutically acceptable salt thereof, when the EGFR gene is found to have a mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation and a L858R mutation as a result of the detection in step (1).
In another aspect, the present invention provides a method of treating and/or preventing a disease, such as a wild-type and/or mutant HER2 kinase-mediated tumor, in a subject, comprising administering to the subject a compound of the present invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20 insymva mutant HER 2.
In particular embodiments, the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
Herein, "HER 2" includes HER2 of a human or non-human mammal. Also, the term "HER 2" includes subtypes.
In the present invention, the HER2 kinase-mediated tumor is preferably a tumor having overexpression of HER2, amplification of HER2 gene, or mutation of HER 2. The "tumor" is not particularly limited, and examples thereof include head and neck cancer, esophageal cancer, gastric cancer, colon cancer, rectal cancer, liver cancer, gallbladder-bile duct cancer, biliary tract cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, cervical cancer, uterine cancer, kidney cancer, bladder cancer, prostate cancer, testicular tumor, bone-soft tissue sarcoma, hematological cancer, multiple myeloma, skin cancer, brain tumor, and mesothelial cancer. Preferred are breast cancer, gastric cancer, esophageal cancer, ovarian cancer, lung cancer, esophageal cancer, gallbladder-bile duct cancer, biliary tract cancer, bladder cancer, and colon cancer, more preferred are breast cancer, gastric cancer, esophageal cancer, biliary tract cancer, ovarian cancer, lung cancer, and esophageal cancer, and further preferred are breast cancer, gastric cancer, and lung cancer.
In the methods of treatment of the present invention, an "effective amount" refers to an amount or dose sufficient to produce the desired therapeutic benefit in an individual in need of such treatment. An effective amount or dose of a compound of the invention can be determined by conventional methods (e.g., modeling, dose escalation, or clinical trials) and by conventional factors (e.g., the mode or route of drug delivery, the pharmacokinetics of the agent, the severity and course of the infection, the health and weight of the individual, and the judgment of the treating physician). Exemplary doses are in the range of about 0.1mg to 1g per day, or about 1mg to 50mg per day, or about 50mg to 250mg per day, or about 250mg to 1g per day. The total dose can be administered as a single dose or as separate dosage units (e.g., BID, TID, QID).
After improvement of the patient's disease occurs, the dosage can be adjusted for prophylactic or maintenance treatment. For example, the dosage or frequency of administration, or both, can be reduced to an amount that maintains the desired therapeutic or prophylactic effect, depending on the symptoms. Of course, if the symptoms have been alleviated to an appropriate extent, treatment may be discontinued. However, when either symptom recurs, the patient may require intermittent treatment for a long period of time. Patients may also require chronic treatment.
Pharmaceutical compositions, formulations and kits
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. Pharmaceutically acceptable carriers, adjuvants, or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
The invention also includes kits (e.g., pharmaceutical packages). The provided kits can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, provided kits may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
The pharmaceutical compositions provided herein may also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over a long period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue for an indefinite period of time, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within the therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to rapidly increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient, e.g., an intramuscular or subcutaneous bolus dose results in a slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, allowing the concentration of the active ingredient in the blood to rise rapidly to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the subject's body. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More generally, however, the compositions are provided in unit dosage form for convenient administration of the precise dosage. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable for the purpose of producing the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired dosage form.
For oral dosages, a representative regimen is one to five oral dosages, particularly two to four oral dosages, typically three oral dosages per day. Using these dosing modes, each dose provides about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing about 0.1 to about 10mg/kg, especially about 1 to about 5 mg/kg.
In order to provide blood levels similar to, or lower than, those used with the injected dose, transdermal doses are generally selected in amounts of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To obtain sufficient steady state levels, a preload bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring and flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream together with, for example, an oil-in-water cream base. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope provided by the present invention.
The compounds of the invention may also be administered by transdermal means. Thus, transdermal administration may be accomplished using a reservoir (reservoir) or porous membrane type, or a variety of solid matrix patches.
The above components of the compositions for oral, injectable or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17th edition,1985, Mack Publishing Company, Easton, Pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6,7 and 8 α -1, 4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substitutions. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Pharmaceutical combination
The compounds of the invention described herein may be used in combination with one or more other active ingredients in pharmaceutical compositions or methods for the treatment of the diseases and conditions described herein. Other additional active ingredients include other therapeutic agents or agents that mitigate the adverse effects of treatment against the intended disease target. The combinations can be used to increase efficacy, ameliorate other disease symptoms, reduce one or more side effects, or reduce the required dosage of the compounds of the invention. The additional active ingredients may be formulated as separate pharmaceutical compositions from the compounds of the present invention or may be included in a single pharmaceutical composition with the compounds of the present invention. The additional active ingredient may be administered simultaneously with, before or after the administration of the compound of the invention.
Combination agents include those active ingredients known or observed to be effective in treating the diseases and conditions described herein, including those effective against another target associated with the disease. For example, the compositions and formulations of the invention, as well as methods of treatment, may further comprise other drugs, such as other agents useful for treating or ameliorating a target disease or associated symptoms or conditions. For cancer indications, the other agents include, but are not limited to, kinase inhibitors, such as EGFR inhibitors (e.g., erlotinib, gefitinib); raf inhibitors (e.g., vemurafenib), VEGFR inhibitors (e.g., sunitinib); standard chemotherapeutic agents, such as alkylating agents, antimetabolites, antitumor antibiotics, topoisomerase inhibitors, platinum drugs, mitotic inhibitors, antibodies, hormonal therapy, or corticosteroids. For pain indications, suitable combination agents include anti-inflammatory agents, such as NSAIDs. The pharmaceutical compositions of the invention may additionally comprise one or more of said active agents, and the method of treatment may additionally comprise administering an effective amount of one or more of said active agents.
Examples
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not specified, in the following examples are generally carried out according to conventional conditions or according to conditions recommended by the manufacturers. Parts and percentages are parts and percentages by weight unless otherwise indicated.
In general, in the preparative schemes, each reaction is carried out in an inert solvent at temperatures ranging from room temperature to reflux temperature (e.g., 0 ℃ to 100 ℃, preferably 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Abbreviations used herein have the following meanings:
POCl 3 : phosphorus oxychloride
DMF: n, N-dimethylformamide
Pd(dppf)Cl 2 :1, 1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
Pd(OAc) 2 Palladium acetate
NBS: n-bromosuccinimide
TEA: triethylamine
TFA: trifluoroacetic acid (trifluoroacetic acid)
Na 2 CO 3 : sodium carbonate
MeOH: methanol
EtOH: ethanol
DCM: methylene dichloride
THF: tetrahydrofuran (THF)
DMF: n, N-dimethylformamide
A Dioxane: dioxane (dioxane)
(Boc) 2 O di-tert-butyl dicarbonate
NaHMDS sodium bis (trimethylsilyl) amide
PPh 3 : triphenylphosphine
Bu 4 NOAc: tetrabutylammonium acetate
In: indium powder
Ti(OEt) 4 : tetraethyl titanate
Allyl bromide: 3-bromopropene
EXAMPLE 1 preparation of N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-1).
Figure BDA0002697128120000431
The following synthetic route was used:
Figure BDA0002697128120000432
step 1. Synthesis of Compound 4-Aminopyrrolo [2,1-f ] [1,2,4] triazine-7-carbaldehyde
Pyrrolo [2,1-f][1,2,4]Triazine-4-amine (10g, 74.6mmol) is dissolved in 150mL DMF, 35mL phosphorus oxychloride is added dropwise in an ice bath, and the temperature is raised to 60 ℃ after the dropwise addition, so as to react overnight. After the reaction, the reaction solution was slowly added to ice water, stirred at room temperature for 30 minutes, adjusted to pH 8 to 9 with 4N NaOH solution, extracted with ethyl acetate (150mL × 3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain 8.65g of pale yellow solid with a yield of 71.5%. ESI-MS: M/z 163[ M ] + +1]。
Step 2, synthesis of compound 4-amino-5-bromopyrrolo [2,1-f ] [1,2,4] triazine-7-formaldehyde
4-amino pyrrolo [2, 1-f)][1,2,4]Triazine-7-carboxaldehyde (8.65g, 53.4mmol) is dissolved in 100mL tetrahydrofuran, cooled to-20 ℃ in an ice bath, dibromohydantoin (9.15g, 32mmol) is added in portions, and the addition is finishedAfter that, the temperature naturally rises to room temperature for reaction overnight. 200mL of water was added to the reaction mixture, and the mixture was stirred at room temperature for 30 minutes, filtered, and the filter cake was washed with water and dried to obtain 8.78g of a pale yellow solid with a yield of 68%. ESI-MS: M/z 243[ M [ ] + +2].
Step 3. Synthesis of Compound 4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazine-7-carbaldehyde
Reacting 4-amino-5-bromopyrrolo [2,1-f ]][1,2,4]Triazine-7-carbaldehyde (8.78g, 36.3mmol), (quinolin-3-yl) boronic acid (7.54g, 43.6mmol), Pd (dppf) Cl 2 (0.8g, 1.09mmol) and sodium carbonate (11.55g, 109mmol) were added to 150mL dioxane and 50mL water, replaced with nitrogen three times, and the temperature was raised to 100 ℃ for 4 hours. The reaction was completed by TLC, the solvent was removed by rotary evaporation from the reaction mixture, and the reaction mixture was diluted with 150mL of water, extracted with ethyl acetate (80mL x 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give a pale yellow solid, 6.5g, and 62% yield. ESI-MS: M/z 290[ M [ ] + +1].
Step 4. Synthesis of Compound (E) -7- (2-methoxyvinyl) -5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-4-amine
(methoxymethyl) triphenylphosphine chloride (15.3g, 44.6mmol) was dissolved in 100mL tetrahydrofuran, NaHMDS (22mL, 44mmol, 2mol/L THF solution) was slowly added dropwise at 0 deg.C, reaction was continued at 0 deg.C for 30 minutes after completion of the addition, and 4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] was slowly added dropwise][1,2,4]After the addition of a solution of triazine-7-carbaldehyde (6.44g, 22.3mmol) in tetrahydrofuran (20mL), the ice bath was removed and the reaction was allowed to proceed at room temperature for 3 hours. TLC detection reaction complete, rotary evaporation to remove solvent, adding 150mL water dilution, using ethyl acetate (80mL x 3) extraction, organic phase with saturated saline water, anhydrous sodium sulfate drying, concentration to obtain brown solid. The solid was added to 30mL ethyl acetate, slurried overnight at room temperature, filtered, and dried to give 5.01g of a pale yellow solid with a yield of 71%. ESI-MS, M/z 318[ M [ ] + +1].
Step 5 Synthesis of Compound 2- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acetaldehyde
Reacting (E) -7- (2-methoxyvinyl) -5- (quinolin-3-yl) pyrrolo [2,1-f][1,2,4]Triazin-4-amine (5.01g, 15.8mmol) was dissolved in 60mL tetrahydrofuran, ice2N hydrochloric acid (20mL) was added dropwise to the bath, and the temperature was raised to 70 ℃ after the addition was completed to react for 2 hours. Cooling the reaction solution to room temperature, adding 100mL of water for dilution, adjusting the pH value to 8-9 by using a 2N NaOH solution, extracting by using ethyl acetate (60mL of 3), washing an organic phase by using saturated saline solution, drying by using anhydrous sodium sulfate, concentrating, and separating by using a silica gel column to obtain a light yellow solid 3.64g with the yield of 76%. ESI-MS: M/z 304[ M [ ] + +1]。
Step 6 Synthesis of Compound N- (1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) -2-methylpropane-2-sulfinamide
2- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) acetaldehyde (3.64g, 12mmol), tert-butylsulfenamide (2.18g, 18mmol) and indium powder (2.07g, 18mmol) were dissolved in 40mL tetrahydrofuran, tetraethyl titanate (5.47g, 24mmol) was slowly added dropwise at 0 ℃ and allowed to naturally warm to room temperature for 2 hours after the addition. The reaction mixture was transferred to an ice bath, 3-bromopropene (3.23g, 26.7mmol) was slowly added dropwise, the ice bath was removed after the addition, and the reaction was allowed to proceed overnight at 60 ℃. The reaction was completed by TLC, the reaction solution was diluted with 120mL of water and 50mL of ethyl acetate, filtered through celite, the aqueous phase was extracted with ethyl acetate (30mL × 3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 1.61g of pale yellow solid with a yield of 30%. ESI-MS, M/z 449M + +1].
Step 7 Synthesis of Compound (tert-butyl 1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) carbamate
Reacting N- (1- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) -2-methylpropane-2-sulfinamide (1.39g, 3.1mmol) is dissolved in 15mL of methanol, 7M methanolic hydrogen chloride (2.2mL, 15.3mmol) is slowly added dropwise at 0 deg.C, reaction is continued at 0 deg.C for 1 hour after completion of the addition, the solvent is removed by rotary evaporation, the residue is dissolved in 20mL of dichloromethane, triethylamine (0.94g, 9.3mmol) is added, and dropwise addition is carried out in ice bath (Boc) 2 O (0.81g, 3.7mmol), after the dropwise addition, the reaction was allowed to warm to room temperature overnight. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and silica gel column separation is carried out to obtain 1.1g of light yellow solid with yield of 80%. ESI-MS, M/z 445[ M ] + +1].
Step 8 Synthesis of Compound (tert-butyl 1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) carbamate
Reacting (1- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) carbamic acid tert-butyl ester (400mg, 0.9mmol) is dissolved in 10mL DMF and NBS (170mg, 0.95mmol) is added portionwise at 0 ℃ and the reaction continued at 0 ℃ for 2h after the addition. The reaction mixture was diluted with 30mL of water, extracted with ethyl acetate (20 mL. x.3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 390mg of a pale yellow solid, 83% yield. ESI-MS: 525[ M/z ] + +2]。
Step 9 Synthesis of Compound (4-amino-6-methylene-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) carbamic acid tert-butyl ester
Reacting (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) carbamic acid tert-butyl ester (390mg, 0.74mmol), tetrabutylammonium acetate (362mg, 1.2mmol), palladium acetate (9mg, 0.04mmol) and triphenylphosphine (39mg, 0.15mmol) were added to 10mL tetrahydrofuran, replaced with nitrogen three times, and the temperature was raised to 90 ℃ for 8 hours. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and light yellow solid 280mg is obtained through silica gel column separation, and the yield is 85%. ESI-MS: M/z 443[ M ═ M + +1].
Step 10 Synthesis of Compound 6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indole-4, 8-diamine
Reacting (4-amino-6-methylene-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4]Triazino [1,6-a]Indol-8-yl) carbamic acid tert-butyl ester (280mg, 0.63mmol) was dissolved in 5mL of ethanol and 1mL of 4N hydrochloric acid and reacted overnight under reflux. 1mL of 4N hydrochloric acid was added, and the reaction was continued under reflux for 6 hours. TLC detection reaction is almost complete, 20mL of water is added for dilution, the pH value is adjusted to 8-9 by saturated sodium carbonate solution, ethyl acetate (20mL of sodium carbonate is used for extraction), an organic phase is washed by saturated saline solution, dried by anhydrous sodium sulfate and concentrated to obtain a light yellow solid 152mg with the yield of 70%. ESI-MS: M/z 343[ M ] + +2]。
Step 11 Synthesis of Compound N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide
Reacting 6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4]Triazino [1,6-a]Indole-4, 8-diamine (152mg, 0.44mmol) and triethylamine (114mg, 0.88mmol) are dissolved in 10mL dichloromethane, the temperature is reduced to-20 ℃ in an ice bath, acryloyl chloride (40mg, 0.44mmol) is slowly dripped, the reaction is continued at-20 ℃ for 1 hour after the dripping is finished, 10mL water is added for dilution, dichloromethane (10mL x 3) is used for extraction, an organic phase is washed by saturated saline water, anhydrous sodium sulfate is dried and concentrated, and light yellow solid 120mg is obtained by silica gel column separation, and the yield is 68%. ESI-MS: M/z: 397[ M [ ] + +1]. 1 H NMR(300MHz,CCl 3 D)δ9.05–8.97(m,1H),8.27(s,1H),8.20(d,J=8.5Hz,1H),7.94–7.77(m,3H),7.66(t,J=7.6Hz,1H),6.31(d,J=16.9Hz,1H),6.04(ddd,J=16.6,10.1,6.0Hz,1H),5.90(dd,J=24.5,8.1Hz,1H),5.64(d,J=10.2Hz,1H),5.60–5.54(m,1H),5.00(d,J=23.5Hz,3H),3.33(q,J=4.2Hz,2H),1.55(s,3H).
Example 2 preparation of (R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-1-a).
Figure BDA0002697128120000461
100mg of racemic compound T-1 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography column and chiral resolution conditions to obtain the objective product T-1-a (retention time: 21.22min, relative content: 40.3%). 1 H NMR(300MHz,CCl 3 D)δ9.05–8.97(m,1H),8.27(s,1H),8.20(d,J=8.5Hz,1H),7.94–7.77(m,3H),7.66(t,J=7.6Hz,1H),6.31(d,J=16.9Hz,1H),6.04(ddd,J=16.6,10.1,6.0Hz,1H),5.90(dd,J=24.5,8.1Hz,1H),5.64(d,J=10.2Hz,1H),5.60–5.54(m,1H),5.00(d,J=23.5Hz,3H),3.33(q,J=4.2Hz,2H),1.55(s,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 60:30:10
Retention time: 21.22 min.
Example 3 preparation of (S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-1-b).
Figure BDA0002697128120000471
100mg of racemic compound T-1 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography column and chiral resolution conditions to obtain the objective product T-1-b (retention time: 26.82min, relative content: 54.6%). 1 H NMR(300MHz,CCl 3 D)δ9.05–8.97(m,1H),8.27(s,1H),8.20(d,J=8.5Hz,1H),7.94–7.77(m,3H),7.66(t,J=7.6Hz,1H),6.31(d,J=16.9Hz,1H),6.04(ddd,J=16.6,10.1,6.0Hz,1H),5.90(dd,J=24.5,8.1Hz,1H),5.64(d,J=10.2Hz,1H),5.60–5.54(m,1H),5.00(d,J=23.5Hz,3H),3.33(q,J=4.2Hz,2H),1.55(s,3H).
Chiral preparative chromatography column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 60:30:10
Retention time: 26.82 min.
Example 4 preparation of N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-2).
Figure BDA0002697128120000481
The following synthetic route was used:
Figure BDA0002697128120000482
step 1 Synthesis of the Compound tert-butyl (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) carbamate
Mixing (4-amino-6-methylene-5- (quinoline-3-yl) -6,7,8, 9-tetrahydro- [1,2, 4-]Triazino [1,6-a ]]Indol-8-yl) carbamic acid tert-butyl ester (305mg, 0.69mmol) was dissolved in 15mL of methanol, 50mg of 10% palladium on carbon was added, replaced with hydrogen three times, and stirred at room temperature overnight under a hydrogen atmosphere of 1 atm. After the reaction is completed, the palladium/carbon is filtered, the filtrate is concentrated, and column chromatography purification is carried out to obtain a light yellow solid of 250mg, wherein the yield is 82%. ESI-MS, M/z 445[ M ] + +1]。
Step 2 Synthesis of Compound 6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indole-4, 8-diamine
Mixing (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2, 4)]Triazino [1,6-a ]]Indol-8-yl) carbamic acid tert-butyl ester (250mg, 0.56mmol) was dissolved in 10mL of dichloromethane, and 3mL of trifluoroacetic acid was added thereto to react at room temperature for 2 hours. And (3) detecting the reaction by TLC (thin layer chromatography), removing the solvent by rotary evaporation, adding 20mL of water for dilution, adjusting the pH to 8-9 by using a saturated sodium carbonate solution, extracting by using dichloromethane (10mL by 3), washing an organic phase by using saturated saline solution, drying by using anhydrous sodium sulfate, and concentrating to obtain a light yellow solid 134mg with the yield of 70%. ESI-MS: M/z 345[ M [ ] + +2]。
Step 3. Synthesis of Compound N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide
Mixing 6-methyl-5- (quinoline-3-yl) -6,7,8, 9-tetrahydro- [1,2, 4-]Triazino [1,6-a ]]Dissolving indole-4, 8-diamine (134mg, 0.39mmol) and triethylamine (79mg, 0.78mmol) in 10mL dichloromethane, cooling to-20 deg.C in ice bath, slowly adding acryloyl chloride (35mg, 0.39mmol) dropwise, continuing to react at-20 deg.C for 1 hr after dropwise addition, adding 10mL water for dilution, extracting with dichloromethane (10mL 3), washing organic phase with 20mL saturated saline solution, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain light yellow solid100mg, yield 65%. ESI-MS, M/z 399[ M ] + +1]. 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
EXAMPLE 5 preparation of (6S,8R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-2-a).
Figure BDA0002697128120000491
100mg of racemic compound T-2 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography and chiral resolution conditions to obtain the objective product T-2-a (retention time: 22.013min, relative content: 22.9%). 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inside diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: MTBE n-Hexane methanol EtOH (0.1% Triethylamine) 45:42:5:8
Retention time: 22.013 min.
Example 6 preparation of (6R,8R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-2-b).
Figure BDA0002697128120000501
100mg of racemic compound T-2 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography column and chiral resolution conditions to obtain the objective product T-2-b (retention time: 24.560min, relative content: 25.4%). 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inside diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: MTBE n-hexane, methanol, ethanol (0.1% triethylamine) 45:42:5:8
Retention time: 24.560 min.
Example 7 preparation of (6R,8S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-2-c).
Figure BDA0002697128120000511
100mg of racemic compound T-2 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography column and chiral resolution conditions to obtain the objective product T-2-c (retention time: 28.840min, relative content: 18.5%). 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: MTBE n-hexane, methanol, ethanol (0.1% triethylamine) 45:42:5:8
Retention time: 28.840 min.
Example 8 preparation of (6S,8S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-2-d).
Figure BDA0002697128120000512
100mg of racemic compound T-2 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative column and chiral resolution conditions to obtain the objective product T-2-d (retention time: 39.140min, relative content: 20.1%). 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inside diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: MTBE n-hexane, methanol, ethanol (0.1% triethylamine) 45:42:5:8
Retention time: 39.140 min.
Example 9 preparation of N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-3).
Figure BDA0002697128120000521
The following synthetic route was used:
Figure BDA0002697128120000522
step 1 Synthesis of Compound (E) -N- (2- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) ethylidene) -2-methylpropane-2-sulfinamide
Reacting 2- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) acetaldehyde (3.09g, 10.2mmol) and tert-butylsulfinamide (1.85g, 15.3mmol) were dissolved in 60mL tetrahydrofuran, and tetraethyltitanate (4.65g, 20.4mmol) was added dropwise slowly at 0 ℃ and allowed to spontaneously rise to room temperature for 2 hours. The reaction was completed by TLC, the reaction solution was diluted with 100mL of water and 100mL of ethyl acetate, filtered through celite, the aqueous phase was extracted with ethyl acetate (40mL × 3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 1.45g of pale yellow solid with a yield of 35%. ESI-MS: M/z 407[ M ═ M + +1].
Step 2 Synthesis of Compound N- (1- (4-amino-5- (quinolin-3-yl)) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) -2-methylpropane-2-sulfinamide
Reacting (E) -N- (2- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) ethylene) -2-methylpropane-2-sulfinamide (1.45g, 3.58mmol) is dissolved in 40mL of dichloromethane, cooled to-20 ℃ and vinyl magnesium bromide (10mL, 1mol/L THF solution) is slowly added dropwiseLiquid), and the reaction is continued at-20 ℃ for 2 hours after the dropwise addition. TLC detected the reaction was complete, 20mL of saturated ammonium chloride solution was added dropwise to the reaction solution and quenched, dichloromethane (20mL x 3) was extracted, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give 1.01g of a pale yellow solid with a yield of 65%. ESI-MS: M/z 435[ M ] + +2]。
Step 3 Synthesis of Compound (tert-butyl 1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) carbamate
Reacting N- (1- (4-amino-5- (quinolin-3-yl)) pyrrolo [2,1-f ]][1,2,4]Triazin-7-yl) but-3-en-2-yl) -2-methylpropane-2-sulfinamide (1.01g, 2.33mmol) is dissolved in 15mL of methanol, 7M methanolic hydrogen chloride (1.7mL, 11.9mmol) is slowly added dropwise at 0 deg.C, reaction is continued at 0 deg.C for 1 hour after completion of the addition, the solvent is removed by rotary evaporation, the residue is dissolved in 20mL of dichloromethane, triethylamine (0.71g, 7mmol) is added, and dropwise addition is carried out in ice bath (Boc) 2 O (0.61g, 2.8mmol), and after the dropwise addition, the reaction was allowed to warm to room temperature overnight. The reaction was completed by TLC detection, and the reaction mixture was concentrated and subjected to silica gel column separation to obtain 0.72g of a pale yellow solid with a yield of 72%. ESI-MS, M/z 431[ M ] + +1].
Step 4 Synthesis of Compound (tert-butyl 1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) carbamate
Reacting (1- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) but-3-en-2-yl) carbamic acid tert-butyl ester (720mg, 1.67mmol) is dissolved in 15mL DMF and NBS (312mg, 1.75mmol) is added portionwise at 0 ℃ and the reaction continued for 2 hours at 0 ℃ after the addition. The reaction mixture was diluted with 50mL of water, extracted with ethyl acetate (20 mL. x.3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 600mg of pale yellow solid with a yield of 70%. ESI-MS, M/z 511[ M ] + +2]。
Step 5 Synthesis of Compound (4-amino-6-methylene-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) carbamic acid Tert-butyl ester
Reacting (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) but-3-en-2-yl) aminomethaneTert-butyl ester (600mg, 1.18mmol), tetrabutylammonium acetate (573mg, 1.9mmol), palladium acetate (13mg, 0.06mmol) and triphenylphosphine (63mg, 0.24mmol) were added to 10mL of tetrahydrofuran, replaced with nitrogen three times, and the temperature was raised to 90 ℃ for reaction for 8 hours. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and light yellow solid 430mg is obtained through silica gel column separation, and the yield is 85%. ESI-MS, M/z 429[ M ] + +1].
Step 6 Synthesis of Compound (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) carbamic acid Tert-butyl ester
Reacting (4-amino-6-methylene-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5]]Pyrrolo [2,1-f][1,2,4]Triazin-7-yl) carbamic acid tert-butyl ester (430mg, 1mmol) was dissolved in 20mL of methanol, 50mg of 10% palladium on carbon was added, replaced with hydrogen three times, and stirred at room temperature under a hydrogen atmosphere of 1 atm overnight. After the reaction is completed, the palladium/carbon is filtered, the filtrate is concentrated, and column chromatography purification is carried out to obtain a light yellow solid 360mg, wherein the yield is 84%. ESI-MS: M/z 431[ M [ ] + +1]。
Step 7. Synthesis of Compound 6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazine-4, 7-diamine
Reacting (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5]]Pyrrolo [2,1-f][1,2,4]Triazin-7-yl) carbamic acid tert-butyl ester (360mg, 0.84mmol) was dissolved in 10mL of dichloromethane, 3mL of trifluoroacetic acid was added, and the reaction was carried out at room temperature for 2 hours. And (3) detecting the reaction by TLC (thin layer chromatography), removing the solvent by rotary evaporation, adding 20mL of water for dilution, adjusting the pH to 8-9 by using a saturated sodium carbonate solution, extracting by using dichloromethane (10mL of sodium carbonate), washing an organic phase by using saturated saline, drying by using anhydrous sodium sulfate, and concentrating to obtain a light yellow solid 205mg with the yield of 74%. ESI-MS: M/z 331[ M [ ] + +2]。
Step 8 Synthesis of Compound N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide
Reacting 6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4, 5%]Pyrrolo [2,1-f][1,2,4]Triazine-4, 7-diamine (205mg, 0.62mmol) and triethylamine (125mg, 1.24mmol) were dissolved in 10mL of dichloromethane,cooling to-20 deg.C in ice bath, slowly adding acryloyl chloride (56mg, 0.62mmol) dropwise, reacting at-20 deg.C for 1 hr, diluting with 20mL water, extracting with dichloromethane (10mL x 3), washing the organic phase with saturated sodium chloride solution, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain light yellow solid 150mg with yield of 63%. ESI-MS: M/z 3385[ M [ ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Example 10 preparation of (6R,7S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-3-a).
Figure BDA0002697128120000551
100mg of racemic compound T-3 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography and chiral resolution conditions to obtain the objective product T-3-a (retention time: 15.52min, relative content: 20.5%). ESI-MS: M/z 3385[ M [ ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 75:10:15
Retention time: 15.52 min.
Example 11 preparation of (6S,7S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-3-b).
Figure BDA0002697128120000561
100mg of racemic compound T-3 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography and chiral resolution conditions to obtain the objective product T-3-b (retention time: 27.54min, relative content: 30.4%). ESI-MS, M/z 3385[ M ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 75:10:15
Retention time: 27.54 min.
Example 12 preparation of (6S,7R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-3-c).
Figure BDA0002697128120000562
Dissolving 100mg of racemic compound T-3 in methanol solution, and separating with chiral preparative chromatographic column and chiral resolution conditionsThe target product T-3-c (retention time: 19.25min, relative content: 18.3%). ESI-MS: M/z 3385[ M [ ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inside diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 75:10:15
Retention time: 19.25 min.
Example 13 preparation of (6R,7R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-3-d).
Figure BDA0002697128120000571
100mg of racemic compound T-3 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative chromatography and chiral resolution conditions to obtain the objective product T-3-d (retention time: 33.22min, relative content: 25.1%). ESI-MS: M/z 3385[ M [ ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 75:10:15
Retention time: 33.22 min.
EXAMPLE 14 preparation of (R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-1-a).
Figure BDA0002697128120000581
The following synthetic route was used:
Figure BDA0002697128120000582
step 1 Synthesis of Compound (R) -N- ((S) -1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) -2-methylpropane-2-sulfinamide
2- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) acetaldehyde (7.2g, 24mmol), (R) -tert-butylsulfinamide (4.36g, 36mmol) and indium powder (4.14g, 36mmol) were dissolved in 60mL tetrahydrofuran, tetraethyltitanate (10.5g, 48mmol) was slowly added dropwise at 0 ℃ and allowed to spontaneously rise to room temperature for 2 hours after the addition. The reaction mixture was transferred to an ice bath, 3-bromopropene (6.46g, 52.4mmol) was slowly added dropwise, the ice bath was removed after the addition, and the reaction was allowed to proceed overnight at 60 ℃. TLC detection reaction is complete, reaction liquid is diluted by adding 200mL water and 150mL ethyl acetate, the diatomite is filtered, aqueous phase is extracted by ethyl acetate (50mL x 3), organic phase is combined, the mixture is washed by saturated saline solution, dried by anhydrous sodium sulfate, concentrated and separated by silica gel column to obtain 3.25g of light yellow solid, and the yield is 29.6%. ESI-MS: M/z 449.1[ M [ ] + +1].
Step 2. Synthesis of Compound (S) - (tert-butyl 1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) carbamate
Reacting (R) -N-((S) -1- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) -2-methylpropane-2-sulfinamide (2.8g, 6.2mmol) is dissolved in 30mL of methanol, 7M methanolic hydrogen chloride solution (4.4mL, 30.6mmol) is slowly added dropwise at 0 deg.C, reaction is continued at 0 deg.C for 1 hour after the addition is complete, the solvent is removed by rotary evaporation, the residue is dissolved in 40mL of dichloromethane, triethylamine (2.0g, 19.0mmol) is added, and dropwise addition is carried out in ice bath (Boc) 2 O (1.62g, 7.4mmol), and after the dropwise addition, the reaction was allowed to warm to room temperature overnight. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and silica gel column separation is carried out to obtain 2.3g of light yellow solid with the yield of 80.6%. ESI-MS, M/z 445.1[ M ] + +1].
Step 3 Synthesis of Compound (S) - (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) pent-4-en-2-yl) carbamic acid tert-butyl ester
Reacting (S) - (1- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) carbamic acid tert-butyl ester (808mg, 1.8mmol) is dissolved in 20mL DMF and NBS (340mg, 1.9mmol) is added portionwise at 0 ℃ and the reaction continued for 2 hours at 0 ℃ after the addition. The reaction mixture was diluted with 50mL of water, extracted with ethyl acetate (30 mL. times.3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 770mg of a pale yellow solid, with a yield of 82.5%. ESI-MS: M/z 525.2[ M [ ] + +1]。
Step 4 Synthesis of Compound (S) - (4-amino-6-methylene-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) carbamic acid tert-butyl ester
Reacting (S) - (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) pent-4-en-2-yl) carbamic acid tert-butyl ester (770mg, 1.46mmol), tetrabutylammonium acetate (720mg, 2.4mmol), palladium acetate (18mg, 0.08mmol) and triphenylphosphine (80mg, 0.3mmol) were added to 20mL tetrahydrofuran, replaced with nitrogen three times, and the temperature was raised to 90 ℃ for reaction for 8 hours. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and light yellow solid 550mg is obtained through silica gel column separation, and the yield is 84.5%. ESI-MS: M/z 443.1[ M ] + +1].
Step 5 Synthesis of Compound (R) -6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indole-4, 8-diamine
Mixing (S) - (4-amino-6-methylene-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4]Triazino [1,6-a]Indol-8-yl) carbamic acid tert-butyl ester (550mg, 1.24mmol) is dissolved in 10mL of ethanol and 4mL of 4N hydrochloric acid and reacted under reflux overnight. TLC detection reaction is complete, 20mL water is added for dilution, the pH value is adjusted to 8-9 by saturated sodium carbonate solution, ethyl acetate (20mL x 3) is used for extraction, an organic phase is washed by saturated saline solution, dried by anhydrous sodium sulfate and concentrated to obtain 300mg of light yellow solid, and the yield is 71.1%. ESI-MS: M/z 343.2[ M [ ] + +1]。
Step 6 Synthesis of Compound (R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide
Mixing (R) -6-methyl-5- (quinolin-3-yl) -8, 9-dihydro- [1,2,4]Triazino [1,6-a]Indole-4, 8-diamine (300mg, 0.88mmol) and triethylamine (228mg, 1.76mmol) are dissolved in 20mL dichloromethane, the temperature is reduced to-20 ℃ in an ice bath, acryloyl chloride (80mg, 0.88mmol) is slowly dripped, the reaction is continuously carried out for 1 hour at-20 ℃ after the dripping is finished, 10mL water is added for dilution, dichloromethane (10mL x 3) is used for extraction, an organic phase is washed by saturated saline solution, anhydrous sodium sulfate is dried and concentrated, 200mg of light yellow solid is obtained by silica gel column separation, and the yield is 61.2%. Chiral purity: 78.6%, and 130mg of off-white solid was obtained by separation under the following chiral preparative column and chiral resolution conditions. ESI-MS: M/z: 397[ M [ ] + +1]. 1 H NMR(300MHz,CDCl 3 )δ9.05–8.97(m,1H),8.27(s,1H),8.20(d,J=8.5Hz,1H),7.94–7.77(m,3H),7.66(t,J=7.6Hz,1H),6.31(d,J=16.9Hz,1H),6.04(ddd,J=16.6,10.1,6.0Hz,1H),5.90(dd,J=24.5,8.1Hz,1H),5.64(d,J=10.2Hz,1H),5.60–5.54(m,1H),5.00(d,J=23.5Hz,3H),3.33(q,J=4.2Hz,2H),1.55(s,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 60:30: 10.
Example 15 preparation of N- ((8S) -4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide (compound T-4) and its stereoisomers (compounds T-4-a and T-4-b).
Figure BDA0002697128120000601
The following synthetic route was used:
Figure BDA0002697128120000611
step 1 Synthesis of the Compound tert-butyl ((8S) -4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) carbamate
Mixing (S) - (4-amino-6-methylene-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4]]Triazino [1,6-a ]]Indol-8-yl) carbamic acid tert-butyl ester (1.0g, 2.25mmol) was dissolved in 25mL of methanol, 200mg of 10% palladium on carbon was added, replaced with hydrogen three times, and stirred at room temperature under a hydrogen atmosphere of 1 atm overnight. After the reaction is completed, the palladium/carbon is filtered, the filtrate is concentrated, and column chromatography purification is carried out to obtain a white-like solid of 850mg, wherein the yield is 84.8%. ESI-MS, M/z 445.1[ M ] + +1]。
Step 2. Synthesis of Compound (8S) -6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indole-4, 8-diamine
(8S) -4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4]Triazino [1,6-a]Indol-8-yl) carbamic acid tert-butyl ester (850mg, 1.91mmol) was dissolved in 25mL of dichloromethane, and 8mL of trifluoroacetic acid was added thereto to react at room temperature for 2 hours. And (3) carrying out TLC detection to complete the reaction, carrying out rotary evaporation to remove the solvent, adding 40mL of water for dilution, adjusting the pH to 8-9 by using saturated sodium carbonate solution, extracting by using dichloromethane (40mL x 3), washing an organic phase by using saturated saline, drying by using anhydrous sodium sulfate, and concentrating to obtain a light yellow solid of 500mg with the yield of 76.1%. ESI-MS: M/z 345.1[ M [ ] + +1]。
Step 3 Synthesis of Compound (8S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide
Mixing (8S) -6-methyl-5- (quinoline-3-yl) -6,7,8, 9-tetrahydro- [1,2, 4-]Triazino [1,6-a]Indole-4, 8-diamine (500mg, 1.45mmol) and triethylamine (360mg, 3.59mmol) were dissolved in 20mL dichloromethane, cooled to-20 ℃ in an ice bath, acryloyl chloride (140mg, 1.6mmol) was slowly added dropwise, and the reaction was continued at-20 ℃ for 1 hour after the addition was completed. Diluting with 10mL of water, extracting with dichloromethane (20 mL. times.3), washing the organic phase with 20mL of saturated brine, drying over anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain 350mg of light yellow solid with yield of 60.6%. ESI-MS, M/z 399[ M ] + +1]. 1 H NMR(400MHz,CCl 3 D)δ9.02(d,J=2.2Hz,1H),8.25–8.14(m,2H),7.94–7.85(m,2H),7.80(ddd,J=8.4,6.8,1.4Hz,1H),7.65(t,J=7.5Hz,1H),6.33(dd,J=17.0,1.4Hz,1H),6.13(dd,J=16.9,10.3Hz,1H),5.77(d,J=8.1Hz,1H),5.68(dd,J=10.2,1.4Hz,1H),5.11(s,2H),4.48(s,1H),3.55(dd,J=15.9,5.5Hz,1H),3.34(s,1H),2.68(ddd,J=16.0,10.5,1.5Hz,1H),2.29(s,1H),1.48–1.37(m,1H),0.79(d,J=6.7Hz,3H).
Step 4 chiral resolution
350mg of (8S) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -6,7,8, 9-tetrahydro- [1,2,4] triazino [1,6-a ] indol-8-yl) acrylamide are dissolved in a methanol solution and subjected to separation under chiral preparative chromatography and chiral resolution conditions described below to give 100mg of an off-white solid (retention time: 25.847min, defined as T-4-a) and 105mg of another type of white solid (retention time: 36.108min, defined as T-4-b).
Chiral separation conditions: chiral preparative chromatography column: CHIRALPAK IC (trade name), 10 mm. times.250 mm (inner diameter. times.length), 5 μm (filler particle diameter)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane methanol ethanol (0.1% diethylamine) 77:8: 15.
Example 16 preparation of N- ((7R) -4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide (compound T-5).
Figure BDA0002697128120000621
The following synthetic route was used:
Figure BDA0002697128120000631
step 1 Synthesis of Compound (R) -N- (2- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) ethylidene) -2-methylpropane-2-sulfinamide
Reacting 2- (4-amino-5- (quinoline-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) acetaldehyde (6.18g, 20.4mmol) and (R) -tert-butylsulfinamide (3.7g, 30.6mmol) were dissolved in 120mL tetrahydrofuran, tetraethyl titanate (9.3g, 40.8mmol) was slowly added dropwise at 0 deg.C, and the reaction was allowed to naturally warm to room temperature for 2 hours after the addition. The reaction was completed by TLC, the reaction solution was diluted with 150mL of water and 150mL of ethyl acetate, filtered through celite, the aqueous phase was extracted with ethyl acetate (80mL × 3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated through a silica gel column to give 3.0g of a pale yellow solid with a yield of 36.1%. ESI-MS, M/z 407.2[ M ] + +1].
Step 2 Synthesis of Compound (R) -N- ((R) -1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) -2-methylpropane-2-sulfinamide
Reacting (R) -N- (2- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) ethylene) -2-methylpropane-2-sulfinamide (2.9g, 6.67mmol) is dissolved in 80mL of dichloromethane, cooled to-20 deg.C, vinyl magnesium bromide (20mL, 1mol/L THF solution) is slowly added dropwise, and the reaction is continued at-20 deg.C for 2 hours after the addition. TLC detection reaction is complete, the reaction solution is dropped into 20mL saturated ammonium chloride solution and quenched, dichloromethane (50mL x 3) is extracted, the organic phase is washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to obtain light yellow solid 2.1g, yield 72.4%. ESI-MS: M/z 435.1[ M [ ] + +2]。
Step 3. Synthesis of Compound (R) - (1- (4-amino-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) carbamic acid tert-butyl ester
Reacting (R) -N- ((R) -1- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) but-3-en-2-yl) -2-methylpropane-2-sulfinamide (2.02g, 4.69mmol) is dissolved in 20mL of methanol, 5M isopropanol hydrochloride solution (10mL) is slowly added dropwise at 0 ℃, the reaction is continued for 1 hour at 0 ℃ after the addition is finished, the solvent is removed by rotary evaporation, the residue is dissolved in 20mL of dichloromethane, triethylamine (1.4g, 14.0mmol) is added, and dropwise addition is carried out under ice bath (Boc) 2 O (1.22g, 5.6mmol), after the dropwise addition, the reaction was allowed to warm to room temperature overnight. The reaction was completed by TLC detection, and the reaction mixture was concentrated and subjected to silica gel column separation to obtain 1.5g of a pale yellow solid with a yield of 74.2%. ESI-MS: M/z 431.1[ M [ ] + +1].
Step 4. Synthesis of Compound (R) - (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) but-3-en-2-yl) carbamic acid tert-butyl ester
Reacting (R) - (1- (4-amino-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) but-3-en-2-yl) carbamic acid tert-butyl ester (1.5g, 3.48mmol) is dissolved in 30mL DMF and NBS (624mg, 3.5mmol) is added portionwise at 0 ℃ and the reaction continued for 2 hours at 0 ℃ after the addition. The reaction mixture was diluted with 50mL of water, extracted with ethyl acetate (40mL × 3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 1.3g of pale yellow solid with a yield of 73.5%. ESI-MS: M/z 509.1[ M [ ] + +2]。
Step 5 Synthesis of Compound (R) - (4-amino-6-methylene-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) carbamic acid Tert-butyl ester
Reacting (R) - (1- (4-amino-6-bromo-5- (quinolin-3-yl) pyrrolo [2, 1-f)][1,2,4]Triazin-7-yl) but-3-en-2-yl) carbamic acid tert-butyl ester (1.3g, 2.56mmol), tetrabutylammonium acetate (1.14g, 3.8mmol), palladium acetate (26mg, 0.12mmol) and triphenylphosphine (126mg, 0.48mmol) were added to 10mL tetrahydrofuran, replaced with nitrogen three times, and the temperature was raised to 90 ℃ for 8 hours. TLC detection shows that the reaction is complete, the reaction solution is concentrated, and the light yellow solid is obtained by separation through a silica gel column with the yield of 850mg and 77.6 percent. ESI-MS: M/z 429.1[ M ] + +1].
Step 6 Synthesis of Compound (7R) - (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) carbamic acid Tert-butyl ester
Reacting (R) - (4-amino-6-methylene-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5]]Pyrrolo [2,1-f][1,2,4]Triazin-7-yl) carbamic acid tert-butyl ester (850mg, 1.98mmol) was dissolved in 20mL methanol, 100mg 10% palladium on carbon was added, hydrogen was substituted three times, and stirring was performed overnight at room temperature under a hydrogen atmosphere of 1 atm. After the reaction is completed, palladium/carbon is filtered, the filtrate is concentrated, and column chromatography purification is carried out to obtain a light yellow solid of 750mg, wherein the yield is 82.3%. ESI-MS, M/z 431.1[ M ] + +1]。
Step 7 Synthesis of Compound (7R) -6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazine-4, 7-diamine
Mixing (7R) - (4-amino-6-methyl-5- (quinoline-3-yl) -7, 8-dihydro-6H-cyclopentano [4,5]Pyrrolo [2,1-f][1,2,4]Triazin-7-yl) carbamic acid tert-butyl ester (750mg, 1.74mmol) was dissolved in 20mL of dichloromethane, 7mL of trifluoroacetic acid was added, and the reaction was carried out at room temperature for 2 hours. And (3) detecting the reaction by TLC (thin layer chromatography), removing the solvent by rotary evaporation, adding 20mL of water for dilution, adjusting the pH to 8-9 by using a saturated sodium carbonate solution, extracting by using dichloromethane (50mL × 3), washing an organic phase by using saturated saline solution, drying by using anhydrous sodium sulfate, and concentrating to obtain a light yellow solid of 420mg, wherein the yield is 73.1%. ESI-MS: M/z 331.1[ M [ ] + +2]。
Step 8 Synthesis of Compound (7R) -N- (4-amino-6-methyl-5- (quinolin-3-yl) -7, 8-dihydro-6H-cyclopenta [4,5] pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) acrylamide
Mixing (7R) -6-methyl-5- (quinoline-3-yl) -7, 8-dihydro-6H-cyclopentano [4, 5%]Pyrrolo [2,1-f][1,2,4]Triazine-4, 7-diamine (420mg, 1.27mmol) and triethylamine (250mg, 2.48mmol) are dissolved in 20mL of dichloromethane, the temperature is reduced to-20 ℃ in an ice bath, acryloyl chloride (112mg, 1.20mmol) is slowly added dropwise, the reaction is continued at-20 ℃ for 1 hour after the dropwise addition, 20mL of water is added for dilution, dichloromethane (10mL & lt3) is used for extraction, the organic phase is washed with saturated saline, dried by anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 300mg of light yellow solid with the yield of 61.5%. ESI-MS: M/z 385.1[ M [ ] + +1]. 1 H NMR(400MHz,CDCl 3 )δ9.04(d,J=2.2Hz,1H),8.25–8.15(m,2H),7.93–7.84(m,2H),7.82(ddd,J=8.3,6.8,1.4Hz,1H),7.66(t,J=7.6Hz,1H),6.31(dd,J=16.9,1.4Hz,1H),6.16(dd,J=16.9,10.1Hz,1H),5.82(d,J=8.3Hz,1H),5.65(d,J=10.2,1H),5.11(s,2H),4.54(s,1H),4.05(m,1H),3.58(s,1H),3.43(m,1H),0.86(d,J=6.7Hz,3H).
Biological activity assay
Biological example 1: kinase inhibition assay
1) EGFR (L858R/T790M) and EGFR (D770_ N771insNPG) kinase activity inhibition assay
The ADP-GloTM Kinase Assay kit (Promega, V9102) is used for determining the inhibitory activity of the drug to be tested on EGFR (L858R/T790M) (SignalChem, E10-122DG-10) and EGFR (D770_ N771insNPG) (SignalChem, E-10-132 GG).
The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3 times of gradient and has 12 concentrations. mu.L of the drug solution was added to each well of a 384 well plate (Perkin Elmer, 6007290) and double wells with 5. mu.L of EGFR (L858R/T790M), 5. mu.L of EGFR (D770-N771 insNPG), respectively. After incubation at 25 ℃ for 15min, 5. mu.L of substrate was added to start the reaction and incubation at 25 ℃ for 60 min. The final reaction concentrations in the system were: 0.5nM EGFR, 10. mu.M ATP,0.03mg/mL Poly (4:1Glu, Tyr) Peptide, HEPES 50mM, EGTA 1mM, MgCl 2 10mM, Brij350.01%. Then 10. mu.L ADP Glo reagent was added and incubation continued for 40min at 25 ℃. After 20. mu.L of the detection reagent was added and incubated at 25 ℃ for 40min, the enzyme inhibition rate of the compound at different concentrations was calculated by reading on an Envision microplate reader (Perkin Elmer, 2104). Data were analyzed using GraphPad Prism 6.0 software, and dose-response curves were derived by fitting the data using non-linear curve regression, and IC was calculated therefrom 50 The value is obtained.
2) HER2(A775_ G776insYVMA) kinase activity inhibition assay
The inhibitory activity of the test drug on HER2(A775_ G776insYVMA) (SignalChem, E27-13BG) was determined using an ADP-GloTM Kinase Assay kit (Promega, V9102).
The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3 times of gradient and has 12 concentrations. To a 384 well plate (Perkin Elmer, 6007290), 0.1. mu.L of the drug solution and 5. mu.L of EGFR (A775_ G7) were added per well76insYVMA), double wells. After incubation at 25 ℃ for 15min, 5. mu.L of substrate was added to start the reaction and incubation at 25 ℃ for 60 min. The final reaction concentrations in the system were: 20nM HER2, 5. mu.M ATP,0.03mg/mL Poly (4:1Glu, Tyr) Peptide, HEPES 50mM, EGTA 1mM, MgCl 2 10mM, Brij350.01%. Then 10. mu.L ADP Glo reagent was added and incubation continued for 40min at 25 ℃. After adding 20. mu.L of detection reagent and incubating at 25 ℃ for 40min, reading on an Envision plate reader (Perkin Elmer,2104) and calculating the inhibition rate of the compound on the enzyme at different concentrations. Data were analyzed using GraphPad Prism 6.0 software, and dose-response curves were derived by fitting the data using non-linear curve regression, and IC was calculated therefrom 50 The value is obtained.
The compounds of the present invention were tested in the above kinase inhibition experiments and found to have potent activity against EGFR (L858R/T790M) and EGFR (D770-N771 insNPG) and HER2 (A775-G776 insYVMA) kinases.
Biological example 2: testing for growth inhibitory Activity against cell lines expressing wild-type and mutant EGFR
1) Test of growth inhibitory Activity of A431 cell, A549 cell, H1975 cell and HCC827 cell
The a431 cells and the a549 cells are wild-type EGFR cells; HCC827 cells are mutant EGFR cells with exon 19 deletion; h1975 cells are EGFR cells with a L858R point mutation and with a T790M point mutation.
The cell concentrations of A431(EGFR) cells, A549 cells (Ex19del), H1975 cells and HCC827 cells were adjusted by adding 50. mu.L of cell suspension to 384-well plates at 37 ℃ with 5% CO, respectively 2 The culture was carried out overnight. The Tecan D300E program was set up. Adding medicine with a Tecan D300E instrument, diluting with 3 times of gradient with the highest concentration of the medicine to be detected being 10 μ M, performing double-hole dilution with 10 concentrations, and continuously culturing for 72 h. The 384 well plates were removed and equilibrated at room temperature for 30min, 30. mu.L of CTG (Promega, G7573) reagent was added to each well, and after 10min at room temperature, the Luminescence values were read on an EnVision (Perkin Elmer 2104) after the signal had stabilized. Inhibition ratio (%) - (1-Lum) Drug to be tested /Lum Negative control ) x100, negative control 0.667% DMSO. IC (integrated circuit) 50 XL-fit software was used for the calculation of (1).
The compounds of the present invention were tested in the cytotoxicity experiments described above, and found to have no inhibitory activity on a431 cells and a549 cells of wild-type EGFR and have potent activity and high selectivity on H1975 cells and HCC827 cells of mutant EGFR, and thus it was found that the compounds of the present invention can inhibit mutant EGFR with exon 19 deletion and mutant EGFR of L858R/T790M with high specificity. The results for representative example compounds are summarized in table 1 below.
Table 1:
Figure BDA0002697128120000671
wherein the structure of TAS6417 is as follows:
Figure BDA0002697128120000672
2)Ba/F 3 parental, Ba/F 3 EGFR-D770-N771ins _ SVD and Ba/F 3 Test of growth inhibitory Activity of HER2-A775_ G776insYVMA cells
Taking cells in logarithmic growth phase, detecting the cell viability by trypan blue exclusion method, and ensuring the cell viability to be more than 90%. Modulation of Ba/F3 parental, Ba/F3 EGFR-D770-N771ins _ SVD and Ba/F 3 The concentration of HER2-A775_ G776insYVMA cells was determined by adding 90. mu.L of cell suspension to a 96-well plate at 37 ℃ with 5% CO 2 The culture was carried out overnight. The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3.16 times of gradient and has 9 concentrations. 10 μ L of the drug solution was added to each well of the 96-well plate, and the culture was continued for 72 hours in triplicate. The 96-well plate was taken out and placed at room temperature for 30min, equal volume of CTG reagent was added to each well, cells were lysed by shaking for 5min on an orbital shaker, and after standing at room temperature for 20min to stabilize luminescence signal, the luminescence value was read on a SpectraMax multi-labeled microplate detector (MD, 2104-0010A). Cell survival (%) — (luminescence of test drug-luminescence of culture control)/(luminescence of cell control-luminescence of culture control) × 100%. Data were analyzed using GraphPad Prism 7.0 software, and dose-effect curves were derived by fitting the data using non-linear sigmoidal regression, and I was calculated therefromC 50 The value is obtained.
The compounds of the invention are directed against Ba/F3 EGFR-D770-N771ins _ SVD and Ba/F 3 HER2-A775_ G776insYVMA cells also have strong activity and high selectivity, and thus it can be seen that the compounds of the present invention can inhibit mutant EGFR with exon 20 insertion with high specificity.
The results for representative example compounds are summarized in table 2 below.
Table 2:
Figure BDA0002697128120000681
biological example 3: pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 per group and compared for pharmacokinetic differences by intravenous or oral administration of a single dose of compound (10 mg/kg oral).
Rats were fed with standard feed and given water. Fasting was initiated 16 hours prior to the test. The drug was dissolved with PEG400 and dimethylsulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L of blood was collected from the orbit into a test tube. The tube contains 30. mu.L of 1% heparin sodium solution. Before use, the tubes were dried overnight at 60 ℃. After completion of blood collection at the last time point, the rats were sacrificed after ether anesthesia.
Immediately after blood collection, the tubes were gently inverted at least 5 times to ensure mixing and then placed on ice. The blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate the plasma from the erythrocytes. Pipette 100 μ L of plasma into a clean plastic centrifuge tube, designating the name of the compound and the time point. Plasma was stored at-80 ℃ before analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on blood drug concentration at different time points for each animal.
Experiments show that the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and treatment effect.
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (13)

1. A compound of formula (IV-3), or a pharmaceutically acceptable salt thereof:
Figure FDF0000017836350000011
wherein the content of the first and second substances,
R 1 is selected from C 1-6 Alkyl, wherein said C 1-6 Alkyl is optionally substituted with one or more D;
r' is selected from H;
R 2 selected from H or D;
R 3 selected from H or D.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
R 1 is C 1-6 An alkyl group;
r' is H;
R 2 is H;
R 3 is H.
3. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, selected from:
Figure FDF0000017836350000012
4. a pharmaceutical composition comprising a compound of any one of claims 1-3, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
5. Use of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 4, for the manufacture of a medicament for the treatment and/or prevention of a mutated EGFR kinase mediated tumor.
6. The use of claim 5, wherein the mutated EGFR is selected from the group consisting of an exon 20 insertion mutant EGFR, an exon 19 deletion mutant EGFR, and an L858R mutant EGFR.
7. The use of claim 6, wherein the exon 20 insertion mutation is selected from the group consisting of V769_ D770insASV, D770_ N771insSVD, D770_ N771insG, H773_ V774insNPH, and H773_ V774 insPH.
8. The use of claim 5 or 6, wherein the mutant EGFR further simultaneously has the T790M mutation.
9. Use of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 4, for the manufacture of a medicament for the treatment and/or prevention of: lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
10. Use of a compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 4, for the manufacture of a medicament for the treatment and/or prevention of a tumor mediated by wild type and/or mutant HER2 kinase.
11. The use of claim 10, wherein the mutated HER2 is selected from the group consisting of ex20insYVMA mutant HER 2.
12. The use of claim 11, wherein said ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
13. Use of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 4, for the manufacture of a medicament for the treatment and/or prevention of: gastric cancer.
CN202011009604.4A 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof Active CN112574208B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210996465.1A CN115181104A (en) 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019109359968 2019-09-29
CN201910935996 2019-09-29

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202210996465.1A Division CN115181104A (en) 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof

Publications (2)

Publication Number Publication Date
CN112574208A CN112574208A (en) 2021-03-30
CN112574208B true CN112574208B (en) 2022-09-13

Family

ID=75120146

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202210996465.1A Pending CN115181104A (en) 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof
CN202011009604.4A Active CN112574208B (en) 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202210996465.1A Pending CN115181104A (en) 2019-09-29 2020-09-23 Substituted fused tricyclic derivatives, compositions and uses thereof

Country Status (2)

Country Link
CN (2) CN115181104A (en)
WO (1) WO2021057796A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115707704A (en) * 2021-08-20 2023-02-21 中山医诺维申新药研发有限公司 Deuterated fused tricyclic compound, and composition and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2861980C (en) * 2012-02-23 2016-11-08 Taiho Pharmaceutical Co., Ltd. Quinolylpyrrolopyrimidyl fused-ring compound or salt thereof
ES2656712T3 (en) * 2013-08-22 2018-02-28 Taiho Pharmaceutical Co., Ltd. Novel quinoline substituted compound
JOP20190073A1 (en) * 2016-10-31 2019-04-07 Taiho Pharmaceutical Co Ltd Selective inhibitor of exon 20 insertion mutant egfr
CN108101905A (en) * 2016-11-24 2018-06-01 中国科学院上海药物研究所 Pyrimido [5,4-b] indolizine or pyrimido [5,4-b] pyrrole biopterin compound, preparation method and the usage
CN110092787B (en) * 2018-01-31 2021-10-15 深圳铂立健医药有限公司 Preparation and application of compound or medicinal salt or composition thereof

Also Published As

Publication number Publication date
CN115181104A (en) 2022-10-14
WO2021057796A1 (en) 2021-04-01
CN112574208A (en) 2021-03-30

Similar Documents

Publication Publication Date Title
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN102918043B (en) As the Pyrrolopyrimidine compounds of CDK4/6 inhibitor
CN102459272B (en) Bicyclic pyrimidine PI3K inhibitor compounds selective for P110 delta, and methods of use
EP2900657B1 (en) Cyclic ether pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
CN110386944B (en) Di (hetero) aryl macrocycles for inhibiting protein kinase activity
CN111285886B (en) Substituted pyrazolo [1,5-a ] pyridine compounds, compositions comprising the same and uses thereof
CN112552302B (en) Substituted aromatic fused ring derivatives, compositions and uses thereof
CN104428299A (en) 5-azaindazole compounds and methods of use
CN112469720A (en) Dual ATM and DNA-PK inhibitors for use in antitumor therapy
KR20230143632A (en) IRAK disintegrant and its uses
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
EP4129987A1 (en) Crystal form of free alkali of nitrogen-containing aromatic derivatives
CN115466273A (en) Substituted alkynyl heterocyclic compounds
CN112574208B (en) Substituted fused tricyclic derivatives, compositions and uses thereof
CN114929707A (en) EED inhibitor and preparation method and application thereof
CN113493439B (en) Substituted acrylamide derivative, composition and application thereof
CN113336760B (en) Substituted amide derivatives, compositions and uses thereof
CN114874189B (en) Substituted heteroaryl derivatives, compositions and uses thereof
JP7323218B2 (en) Substituted Fused Aromatic Ring Derivatives, Compositions Thereof, and Uses Thereof
CN111349084B (en) Aminopyrimidines useful for inhibiting protein kinase activity
CA3116141C (en) Cycloalkane-1,3-diamine derivative
KR20230057380A (en) 1H-imidazo[4,5-H]quinazoline compounds as novel selective FLT3 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant